1
|
Butler C, Dunmire M, Choi J, Szalai G, Johnson A, Lei W, Chen X, Liu L, Li W, Walter MJ, Liu T. HSPA9/mortalin inhibition disrupts erythroid maturation through a TP53-dependent mechanism in human CD34+ hematopoietic progenitor cells. Cell Stress Chaperones 2024; 29:300-311. [PMID: 38508444 PMCID: PMC10998001 DOI: 10.1016/j.cstres.2024.03.006] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2023] [Revised: 03/16/2024] [Accepted: 03/16/2024] [Indexed: 03/22/2024] Open
Abstract
Myelodysplastic syndromes (MDS) are a heterogeneous group of clonal hematopoietic stem cell malignancies characterized by abnormal hematopoietic cell maturation, increased apoptosis of bone marrow cells, and anemia. They are the most common myeloid blood cancers in American adults. The full complement of gene mutations that contribute to the phenotypes or clinical symptoms in MDS is not fully understood. Around 10%-25% of MDS patients harbor an interstitial heterozygous deletion on the long arm of chromosome 5 [del(5q)], creating haploinsufficiency for a large set of genes, including HSPA9. The HSPA9 gene encodes for the protein mortalin, a highly conserved heat shock protein predominantly localized in mitochondria. Our prior study showed that knockdown of HSPA9 induces TP53-dependent apoptosis in human CD34+ hematopoietic progenitor cells. In this study, we explored the role of HSPA9 in regulating erythroid maturation using human CD34+ cells. We inhibited the expression of HSPA9 using gene knockdown and pharmacological inhibition and found that inhibition of HSPA9 disrupted erythroid maturation as well as increased expression of p53 in CD34+ cells. To test whether the molecular mechanism of HSPA9 regulating erythroid maturation is TP53-dependent, we knocked down HSPA9 and TP53 individually or in combination in human CD34+ cells. We found that the knockdown of TP53 partially rescued the erythroid maturation defect induced by HSPA9 knockdown, suggesting that the defect in cells with reduced HSPA9 expression is TP53-dependent. Collectively, these findings indicate that reduced levels of HSPA9 may contribute to the anemia observed in del(5q)-associated MDS patients due to the activation of TP53.
Collapse
Affiliation(s)
- Christopher Butler
- Department of Biomedical Sciences, West Virginia School of Osteopathic Medicine, Lewisburg, WV, USA
| | - Morgan Dunmire
- Department of Biomedical Sciences, West Virginia School of Osteopathic Medicine, Lewisburg, WV, USA
| | - Jaebok Choi
- Department of Medicine, Division of Oncology, Washington University School of Medicine, St. Louis, MO, USA
| | - Gabor Szalai
- Department of Biomedical Sciences, Burrell College of Osteopathic Medicine, Las Cruces, NM, USA
| | - Anissa Johnson
- Department of Biomedical Sciences, Burrell College of Osteopathic Medicine, Las Cruces, NM, USA
| | - Wei Lei
- Department of Pharmaceutical and Graduate Life Sciences, Manchester University College of Pharmacy, Natural and Health Sciences, Fort Wayne, IN, USA
| | - Xin Chen
- Department of Pharmaceutical and Clinical Sciences, College of Pharmacy and Health Sciences, Campbell University, Buies Creek, NC, USA
| | - Liang Liu
- Department of Cancer Biology, Wake Forest School of Medicine, Winston-Salem, NC, USA
| | - Wei Li
- Department of Biomedical Sciences, Joan C. Edwards School of Medicine, Marshall University, Huntington, WV, USA
| | - Matthew J Walter
- Department of Medicine, Division of Oncology, Washington University School of Medicine, St. Louis, MO, USA
| | - Tuoen Liu
- Department of Biomedical Sciences, West Virginia School of Osteopathic Medicine, Lewisburg, WV, USA.
| |
Collapse
|
2
|
Chang X, Ji C, Zhang T, Huang H. Prenatal to preimplantation genetic diagnosis of a novel compound heterozygous mutation in HSPA9 associated with Even-Plus syndrome. Clin Chim Acta 2024; 555:117803. [PMID: 38281662 DOI: 10.1016/j.cca.2024.117803] [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: 12/24/2023] [Revised: 01/21/2024] [Accepted: 01/23/2024] [Indexed: 01/30/2024]
Abstract
BACKGROUND Heat shock protein family A member 9 (HSPA9) prevents unfolded and dysfunctional protein accumulation, with genetic variants known to be pathogenic. Here, we determined the genetic cause of Even-Plus syndrome (OMIM: 616854) in a Chinese family. METHODS We collected samples from two affected and two normal individuals. Whole-exome sequencing was performed to identify their genetic profiles. Potential variants were validated using Sanger sequencing. Assisted reproduction with mutation-free embryos successfully blocked the transmission of mutations. RESULTS We identified novel inherited pathogenic complex heterozygous variations in the HSPA9 gene in the two affected fetuses. Three-dimensional spatial simulation of the HSPA9 protein after prediction of the mutated RNA splicing pattern abolished part of the substrate-binding domain of the protein. According to ACMG guidelines, c. 1822-1G>A and c. 1411-3T>G were classified as pathogenic and likely pathogenic, respectively. Mutation-free embryos were selected for transplantation and reconfirmed to possess no mutations. A healthy daughter was successfully born into the family. CONCLUSIONS This study is the first to report complex heterozygous variations in the HSPA9 gene that influence alternative splicing in early pregnancy. Our findings expand on the mutational spectrum leading to Even-Plus syndrome and provide a basis for genetic counseling and future embryonic studies.
Collapse
Affiliation(s)
- Xiaoxia Chang
- Department of Pediatrics, The First Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu Province, China; Department of Obstetrics and Gynecology, the First Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu Province, China
| | - Chunmin Ji
- Department of Obstetrics and Gynecology, Air Force Hospital of Eastern Theater, Nanjing, Jiangsu Province, China
| | - Ting Zhang
- Department of Obstetrics and Gynecology, the First Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu Province, China
| | - Huan Huang
- Department of Pediatrics, The First Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu Province, China; Department of Obstetrics and Gynecology, the First Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu Province, China.
| |
Collapse
|
3
|
Wu M, Xu J, Zhang Y, Wen Z. Learning from Zebrafish Hematopoiesis. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2023; 1442:137-157. [PMID: 38228963 DOI: 10.1007/978-981-99-7471-9_9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/18/2024]
Abstract
Hematopoiesis is a complex process that tightly regulates the generation, proliferation, differentiation, and maintenance of hematopoietic cells. Disruptions in hematopoiesis can lead to various diseases affecting both hematopoietic and non-hematopoietic systems, such as leukemia, anemia, thrombocytopenia, rheumatoid arthritis, and chronic granuloma. The zebrafish serves as a powerful vertebrate model for studying hematopoiesis, offering valuable insights into both hematopoietic regulation and hematopoietic diseases. In this chapter, we present a comprehensive overview of zebrafish hematopoiesis, highlighting its distinctive characteristics in hematopoietic processes. We discuss the ontogeny and modulation of both primitive and definitive hematopoiesis, as well as the microenvironment that supports hematopoietic stem/progenitor cells. Additionally, we explore the utility of zebrafish as a disease model and its potential in drug discovery, which not only advances our understanding of the regulatory mechanisms underlying hematopoiesis but also facilitates the exploration of novel therapeutic strategies for hematopoietic diseases.
Collapse
Affiliation(s)
- Mei Wu
- Affiliated Hospital of Guangdong Medical University and Key Laboratory of Zebrafish Model for Development and Disease of Guangdong Medical University, Zhanjiang, Guangdong, China
| | - Jin Xu
- South China University of Technology, School of Medicine, Guangzhou, Guangdong, China.
| | - Yiyue Zhang
- South China University of Technology, School of Medicine, Guangzhou, Guangdong, China.
| | - Zilong Wen
- Southern University of Science and Technology, School of Life Sciences, Shenzhen, Guangdong, China.
| |
Collapse
|
4
|
Yi ZN, Chen XK, Ma ACH. Modeling leukemia with zebrafish (Danio rerio): Towards precision medicine. Exp Cell Res 2022; 421:113401. [PMID: 36306826 DOI: 10.1016/j.yexcr.2022.113401] [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: 07/06/2022] [Revised: 10/06/2022] [Accepted: 10/20/2022] [Indexed: 12/29/2022]
Abstract
Leukemia is a type of blood cancer characterized by high genetic heterogeneity and fatality. While chemotherapy remains the primary form of treatment for leukemia, its effectiveness was profoundly diminished by the genetic heterogeneity and cytogenetic abnormalities of leukemic cells. Therefore, there is an unmet need to develop precision medicine for leukemia with distinct genetic backgrounds. Zebrafish (Danio rerio), a freshwater fish with exceptional feasibility in genome editing, is a powerful tool for rapid human cancer modeling. In the past decades, zebrafish have been adopted in modeling human leukemia, exploring the molecular mechanisms of underlying genetic abnormalities, and discovering novel therapeutic agents. Although many recurrent mutations of leukemia have been modeled in zebrafish for pathological study and drug discovery, its great potential in leukemia modeling was not yet fully exploited, particularly in precision medicine. In this review, we evaluated the current zebrafish models of leukemia/pre-leukemia and genetic techniques and discussed the potential of zebrafish models with novel techniques, which may contribute to the development of zebrafish as a disease model for precision medicine in treating leukemia.
Collapse
Affiliation(s)
- Zhen-Ni Yi
- Department of Health Technology and Informatics, The Hong Kong Polytechnic University, Hong Kong, China
| | - Xiang-Ke Chen
- Department of Health Technology and Informatics, The Hong Kong Polytechnic University, Hong Kong, China
| | - Alvin Chun-Hang Ma
- Department of Health Technology and Informatics, The Hong Kong Polytechnic University, Hong Kong, China.
| |
Collapse
|
5
|
Mathangasinghe Y, Fauvet B, Jane SM, Goloubinoff P, Nillegoda NB. The Hsp70 chaperone system: distinct roles in erythrocyte formation and maintenance. Haematologica 2021; 106:1519-1534. [PMID: 33832207 PMCID: PMC8168490 DOI: 10.3324/haematol.2019.233056] [Citation(s) in RCA: 19] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2019] [Indexed: 01/14/2023] Open
Abstract
Erythropoiesis is a tightly regulated cell differentiation process in which specialized oxygen- and carbon dioxide-carrying red blood cells are generated in vertebrates. Extensive reorganization and depletion of the erythroblast proteome leading to the deterioration of general cellular protein quality control pathways and rapid hemoglobin biogenesis rates could generate misfolded/aggregated proteins and trigger proteotoxic stresses during erythropoiesis. Such cytotoxic conditions could prevent proper cell differentiation resulting in premature apoptosis of erythroblasts (ineffective erythropoiesis). The heat shock protein 70 (Hsp70) molecular chaperone system supports a plethora of functions that help maintain cellular protein homeostasis (proteostasis) and promote red blood cell differentiation and survival. Recent findings show that abnormalities in the expression, localization and function of the members of this chaperone system are linked to ineffective erythropoiesis in multiple hematological diseases in humans. In this review, we present latest advances in our understanding of the distinct functions of this chaperone system in differentiating erythroblasts and terminally differentiated mature erythrocytes. We present new insights into the protein repair-only function(s) of the Hsp70 system, perhaps to minimize protein degradation in mature erythrocytes to warrant their optimal function and survival in the vasculature under healthy conditions. The work also discusses the modulatory roles of this chaperone system in a wide range of hematological diseases and the therapeutic gain of targeting Hsp70.
Collapse
Affiliation(s)
| | - Bruno Fauvet
- Department of Plant Molecular Biology, Lausanne University, Lausanne
| | - Stephen M Jane
- Central Clinical School, Monash University, Prahran, Victoria, Australia; Department of Hematology, Alfred Hospital, Monash University, Prahran, Victoria
| | | | - Nadinath B Nillegoda
- Australian Regenerative Medicine Institute, Monash University, Clayton, Victoria.
| |
Collapse
|
6
|
Li S, Zheng X, Wang T, Xue J. Clinical characterization of novel HSPA9 splice variant in a Chinese woman. Clin Genet 2021; 99:609-610. [PMID: 33398880 DOI: 10.1111/cge.13910] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2020] [Revised: 12/24/2020] [Accepted: 12/26/2020] [Indexed: 11/30/2022]
Affiliation(s)
- Shufeng Li
- Department of Biochemistry and Molecular Biology, Medical School of Southeast University, Nanjing, China
| | - Xiaojuan Zheng
- Department of Biochemistry and Molecular Biology, Medical School of Southeast University, Nanjing, China
| | - Ting Wang
- Department of Biochemistry and Molecular Biology, Medical School of Southeast University, Nanjing, China
| | - Jun Xue
- Department of Hematology, Nanjing First Hospital, Nanjing Medical University, Nanjing, China
| |
Collapse
|
7
|
Sen T, Jain M, Gram M, Mattebo A, Soneji S, Walkley CR, Singbrant S. Enhancing mitochondrial function in vivo rescues MDS-like anemia induced by pRb deficiency. Exp Hematol 2020; 88:28-41. [PMID: 32629063 DOI: 10.1016/j.exphem.2020.06.006] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2020] [Revised: 06/25/2020] [Accepted: 06/30/2020] [Indexed: 12/14/2022]
Abstract
Erythropoiesis is intimately coupled to cell division, and deletion of the cell cycle regulator retinoblastoma protein (pRb) causes anemia in mice. Erythroid-specific deletion of pRb has been found to result in inefficient erythropoiesis because of deregulated coordination of cell cycle exit and mitochondrial biogenesis. However, the pathophysiology remains to be fully described, and further characterization of the link between cell cycle regulation and mitochondrial function is needed. To this end we further assessed conditional erythroid-specific deletion of pRb. This resulted in macrocytic anemia, despite elevated levels of erythropoietin (Epo), and an accumulation of erythroid progenitors in the bone marrow, a phenotype strongly resembling refractory anemia associated with myelodysplastic syndromes (MDS). Using high-fractionation fluorescence-activated cell sorting analysis for improved phenotypic characterization, we illustrate that erythroid differentiation was disrupted at the orthochromatic stage. Transcriptional profiling of sequential purified populations revealed failure to upregulate genes critical for mitochondrial function such as Pgc1β, Alas2, and Abcb7 specifically at the block, together with disturbed heme production and iron transport. Notably, deregulated ABCB7 causes ring sideroblastic anemia in MDS patients, and the mitochondrial co-activator PGC1β is heterozygously lost in del5q MDS. Importantly, the anemia could be rescued through enhanced PPAR signaling in vivo via either overexpression of Pgc1β or bezafibrate administration. In conclusion, lack of pRb results in MDS-like anemia with disrupted differentiation and impaired mitochondrial function at the orthochromatic erythroblast stage. Our findings reveal for the first time a role for pRb in heme and iron regulation, and indicate that pRb-induced anemia can be rescued in vivo through therapeutic enhancement of PPAR signaling.
Collapse
Affiliation(s)
- Taha Sen
- Division of Molecular Medicine and Gene Therapy, Lund Stem Cell Center, Lund University, Lund, Sweden
| | - Mayur Jain
- Division of Molecular Medicine and Gene Therapy, Lund Stem Cell Center, Lund University, Lund, Sweden
| | - Magnus Gram
- Department of Clinical Sciences Lund, Pediatrics, Lund University, Skane University Hospital Lund, Lund, Sweden
| | - Alexander Mattebo
- Division of Molecular Medicine and Gene Therapy, Lund Stem Cell Center, Lund University, Lund, Sweden
| | - Shamit Soneji
- Division of Molecular Hematology, Lund Stem Cell Center, Lund University, Lund, Sweden
| | - Carl R Walkley
- St. Vincent's Institute of Medical Research and Department of Medicine, University of Melbourne, Fitzroy, VIC, Australia
| | - Sofie Singbrant
- Division of Molecular Medicine and Gene Therapy, Lund Stem Cell Center, Lund University, Lund, Sweden
| |
Collapse
|
8
|
Chua BA, Van Der Werf I, Jamieson C, Signer RAJ. Post-Transcriptional Regulation of Homeostatic, Stressed, and Malignant Stem Cells. Cell Stem Cell 2020; 26:138-159. [PMID: 32032524 PMCID: PMC7158223 DOI: 10.1016/j.stem.2020.01.005] [Citation(s) in RCA: 45] [Impact Index Per Article: 11.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
Cellular identity is not driven by differences in genomic content but rather by epigenomic, transcriptomic, and proteomic heterogeneity. Although regulation of the epigenome plays a key role in shaping stem cell hierarchies, differential expression of transcripts only partially explains protein abundance. The epitranscriptome, translational control, and protein degradation have emerged as fundamental regulators of proteome complexity that regulate stem cell identity and function. Here, we discuss how post-transcriptional mechanisms enable stem cell homeostasis and responsiveness to developmental cues and environmental stressors by rapidly shaping the content of their proteome and how these processes are disrupted in pre-malignant and malignant states.
Collapse
Affiliation(s)
- Bernadette A Chua
- Division of Regenerative Medicine, Department of Medicine, Moores Cancer Center, University of California San Diego, La Jolla, CA, 92093 USA
| | - Inge Van Der Werf
- Division of Regenerative Medicine, Department of Medicine, Moores Cancer Center, University of California San Diego, La Jolla, CA, 92093 USA; Sanford Stem Cell Clinical Center, La Jolla, CA 92037, USA
| | - Catriona Jamieson
- Division of Regenerative Medicine, Department of Medicine, Moores Cancer Center, University of California San Diego, La Jolla, CA, 92093 USA; Sanford Stem Cell Clinical Center, La Jolla, CA 92037, USA.
| | - Robert A J Signer
- Division of Regenerative Medicine, Department of Medicine, Moores Cancer Center, University of California San Diego, La Jolla, CA, 92093 USA.
| |
Collapse
|
9
|
Badaat I, Mirza S, Padron E, Sallman D, Komrokji R, Song J, Hussaini MO. Concurrent mutations in other epigenetic modulators portend better prognosis in BCOR-mutated myelodysplastic syndrome. J Clin Pathol 2019; 73:209-212. [PMID: 31771970 DOI: 10.1136/jclinpath-2019-206132] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2019] [Revised: 10/04/2019] [Accepted: 10/14/2019] [Indexed: 12/21/2022]
Abstract
INTRODUCTION The role of single mutations has been extensively studied myelodysplastic syndromes (MDS), but the impact of genetic aberrations in the context of other mutations is less well understood. BCOR is an epigenetic transcriptional corepressor. In MDS, BCOR mutations are rare and certain mutations are associated with poor prognosis. Our aim was to investigate the role of concurrent mutations in epigenetic MDS-driver genes in BCOR-mutated MDS. We hypothesised that these would be redundant and would not contribute to worse prognosis. METHODS Internal Next Generation Sequencing (NGS) database with targeted genetic profiling of >4000 tumor cases was queried to locate cases of MDS with BCL6 Corepressor protein (BCOR) mutations only (pBCOR) and cBCOR (comutated epigenetic modulators: TET2, ASXL1, DNMT3A, EZH2, IDH2, IDH1, BCORL1, ATRX). Overall survival was determined by chart review. Fischer's exact test and unpaired t-test was performed for statistical analysis. RESULTS 25 patients with cBCOR were detected. Only five MDS patients with pBCOR were found. The number of patients with comutations (cBCOR) in epigenetic modulators comprised TET2 (n=5), ASXL1 (n=9), DNMT3A (n=11), EZH2 (n=2), IDH2 (n=4), IDH1 (n=1), BCORL1 (n=3), ATRX (n=1). cBCOR overall survival was 23.8 months versus 11.8 months for pBCOR group. CONCLUSIONS In this study, we confirm the rarity of BCOR mutations. Our results show that there is a trend towards poorer prognosis in patient with pBCOR versus cBCOR although statistical significance was not reached. This may be due to enrichment of poor cytogenetics in pBCOR or increased responsiveness to hypomethylating agents in cBCOR. Larger studies are needed to validate our data.
Collapse
Affiliation(s)
- Ibraahim Badaat
- College of Arts and Sciences, University of South Florida, Tampa, Florida, USA
| | - Sabbir Mirza
- College of Arts and Sciences, University of South Florida, Tampa, Florida, USA
| | - Eric Padron
- Division of Malignant Hematology, H Lee Moffitt Cancer Center and Research Institute, Tampa, Florida, USA
| | - David Sallman
- Division of Malignant Hematology, H Lee Moffitt Cancer Center and Research Institute, Tampa, Florida, USA
| | - Rami Komrokji
- Division of Malignant Hematology, H Lee Moffitt Cancer Center and Research Institute, Tampa, Florida, USA
| | - Jinming Song
- Hematopathology, H Lee Moffitt Cancer Center and Research Institute, Tampa, Florida, USA
| | - Mohammad O Hussaini
- Hematopathology, H Lee Moffitt Cancer Center and Research Institute, Tampa, Florida, USA
| |
Collapse
|
10
|
Heat shock protein 60 regulates yolk sac erythropoiesis in mice. Cell Death Dis 2019; 10:766. [PMID: 31601784 PMCID: PMC6786998 DOI: 10.1038/s41419-019-2014-2] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2019] [Revised: 09/22/2019] [Accepted: 09/26/2019] [Indexed: 12/14/2022]
Abstract
The yolk sac is the first site of blood-cell production during embryonic development in both murine and human. Heat shock proteins (HSPs), including HSP70 and HSP27, have been shown to play regulatory roles during erythropoiesis. However, it remains unknown whether HSP60, a molecular chaperone that resides mainly in mitochondria, could also regulate early erythropoiesis. In this study, we used Tie2-Cre to deactivate the Hspd1 gene in both hematopoietic and vascular endothelial cells, and found that Tie2-Cre+Hspd1f/f (HSP60CKO) mice were embryonic lethal between the embryonic day 10.5 (E10.5) and E11.5, exhibiting growth retardation, anemia, and vascular defects. Of these, anemia was observed first, independently of vascular and growth phenotypes. Reduced numbers of erythrocytes, as well as an increase in cell apoptosis, were found in the HSP60CKO yolk sac as early as E9.0, indicating that deletion of HSP60 led to abnormality in yolk sac erythropoiesis. Deletion of HSP60 was also able to reduce mitochondrial membrane potential and the expression of the voltage-dependent anion channel (VDAC) in yolk sac erythrocytes. Furthermore, cyclosporine A (CsA), which is a well-recognized modulator in regulating the opening of the mitochondrial permeability transition pore (mPTP) by interacting with Cyclophilin D (CypD), could significantly decrease cell apoptosis and partially restore VDAC expression in mutant yolk sac erythrocytes. Taken together, we demonstrated an essential role of HSP60 in regulating yolk sac cell survival partially via a mPTP-dependent mechanism.
Collapse
|
11
|
Konantz M, Schürch C, Hanns P, Müller JS, Sauteur L, Lengerke C. Modeling hematopoietic disorders in zebrafish. Dis Model Mech 2019; 12:12/9/dmm040360. [PMID: 31519693 PMCID: PMC6765189 DOI: 10.1242/dmm.040360] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022] Open
Abstract
Zebrafish offer a powerful vertebrate model for studies of development and disease. The major advantages of this model include the possibilities of conducting reverse and forward genetic screens and of observing cellular processes by in vivo imaging of single cells. Moreover, pathways regulating blood development are highly conserved between zebrafish and mammals, and several discoveries made in fish were later translated to murine and human models. This review and accompanying poster provide an overview of zebrafish hematopoiesis and discuss the existing zebrafish models of blood disorders, such as myeloid and lymphoid malignancies, bone marrow failure syndromes and immunodeficiencies, with a focus on how these models were generated and how they can be applied for translational research. Summary: This At A Glance article and poster summarize the last 20 years of research in zebrafish models for hematopoietic disorders, highlighting how these models were created and are being applied for translational research.
Collapse
Affiliation(s)
- Martina Konantz
- Department of Biomedicine, University of Basel and University Hospital Basel, Basel 4031, Switzerland
| | - Christoph Schürch
- Department of Biomedicine, University of Basel and University Hospital Basel, Basel 4031, Switzerland
| | - Pauline Hanns
- Department of Biomedicine, University of Basel and University Hospital Basel, Basel 4031, Switzerland
| | - Joëlle S Müller
- Department of Biomedicine, University of Basel and University Hospital Basel, Basel 4031, Switzerland
| | - Loïc Sauteur
- Department of Biomedicine, University of Basel and University Hospital Basel, Basel 4031, Switzerland
| | - Claudia Lengerke
- Department of Biomedicine, University of Basel and University Hospital Basel, Basel 4031, Switzerland.,Division of Hematology, University of Basel and University Hospital Basel, Basel 4031, Switzerland
| |
Collapse
|
12
|
Leitch HA, Gattermann N. Hematologic improvement with iron chelation therapy in myelodysplastic syndromes: Clinical data, potential mechanisms, and outstanding questions. Crit Rev Oncol Hematol 2019; 141:54-72. [DOI: 10.1016/j.critrevonc.2019.06.002] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2018] [Revised: 11/25/2018] [Accepted: 06/03/2019] [Indexed: 12/25/2022] Open
|
13
|
Congenital sideroblastic anemia: Advances in gene mutations and pathophysiology. Gene 2018; 668:182-189. [DOI: 10.1016/j.gene.2018.05.074] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2018] [Accepted: 05/18/2018] [Indexed: 01/19/2023]
|
14
|
Clinical and genetic aspects of defects in the mitochondrial iron-sulfur cluster synthesis pathway. J Biol Inorg Chem 2018; 23:495-506. [PMID: 29623423 PMCID: PMC6006192 DOI: 10.1007/s00775-018-1550-z] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2017] [Accepted: 02/26/2018] [Indexed: 12/12/2022]
Abstract
Iron-sulfur clusters are evolutionarily conserved biological structures which play an important role as cofactor for multiple enzymes in eukaryotic cells. The biosynthesis pathways of the iron-sulfur clusters are located in the mitochondria and in the cytosol. The mitochondrial iron-sulfur cluster biosynthesis pathway (ISC) can be divided into at least twenty enzymatic steps. Since the description of frataxin deficiency as the cause of Friedreich's ataxia, multiple other deficiencies in ISC biosynthesis pathway have been reported. In this paper, an overview is given of the clinical, biochemical and genetic aspects reported in humans affected by a defect in iron-sulfur cluster biosynthesis.
Collapse
|
15
|
Gore AV, Pillay LM, Venero Galanternik M, Weinstein BM. The zebrafish: A fintastic model for hematopoietic development and disease. WILEY INTERDISCIPLINARY REVIEWS-DEVELOPMENTAL BIOLOGY 2018; 7:e312. [PMID: 29436122 DOI: 10.1002/wdev.312] [Citation(s) in RCA: 122] [Impact Index Per Article: 20.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/28/2017] [Revised: 11/30/2017] [Accepted: 12/03/2017] [Indexed: 12/19/2022]
Abstract
Hematopoiesis is a complex process with a variety of different signaling pathways influencing every step of blood cell formation from the earliest precursors to final differentiated blood cell types. Formation of blood cells is crucial for survival. Blood cells carry oxygen, promote organ development and protect organs in different pathological conditions. Hematopoietic stem and progenitor cells (HSPCs) are responsible for generating all adult differentiated blood cells. Defects in HSPCs or their downstream lineages can lead to anemia and other hematological disorders including leukemia. The zebrafish has recently emerged as a powerful vertebrate model system to study hematopoiesis. The developmental processes and molecular mechanisms involved in zebrafish hematopoiesis are conserved with higher vertebrates, and the genetic and experimental accessibility of the fish and the optical transparency of its embryos and larvae make it ideal for in vivo analysis of hematopoietic development. Defects in zebrafish hematopoiesis reliably phenocopy human blood disorders, making it a highly attractive model system to screen small molecules to design therapeutic strategies. In this review, we summarize the key developmental processes and molecular mechanisms of zebrafish hematopoiesis. We also discuss recent findings highlighting the strengths of zebrafish as a model system for drug discovery against hematopoietic disorders. This article is categorized under: Adult Stem Cells, Tissue Renewal, and Regeneration > Stem Cell Differentiation and Reversion Vertebrate Organogenesis > Musculoskeletal and Vascular Nervous System Development > Vertebrates: Regional Development Comparative Development and Evolution > Organ System Comparisons Between Species.
Collapse
Affiliation(s)
- Aniket V Gore
- Division of Developmental Biology, Eunice Kennedy Shriver National Institute of Child Health and Human Development, NIH, Bethesda, Maryland
| | - Laura M Pillay
- Division of Developmental Biology, Eunice Kennedy Shriver National Institute of Child Health and Human Development, NIH, Bethesda, Maryland
| | - Marina Venero Galanternik
- Division of Developmental Biology, Eunice Kennedy Shriver National Institute of Child Health and Human Development, NIH, Bethesda, Maryland
| | - Brant M Weinstein
- Division of Developmental Biology, Eunice Kennedy Shriver National Institute of Child Health and Human Development, NIH, Bethesda, Maryland
| |
Collapse
|
16
|
Wachnowsky C, Liu Y, Yoon T, Cowan JA. Regulation of human Nfu activity in Fe-S cluster delivery-characterization of the interaction between Nfu and the HSPA9/Hsc20 chaperone complex. FEBS J 2017; 285:391-410. [PMID: 29211945 DOI: 10.1111/febs.14353] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2017] [Revised: 10/24/2017] [Accepted: 12/01/2017] [Indexed: 12/11/2022]
Abstract
Iron-sulfur cluster biogenesis is a complex, but highly regulated process that involves de novo cluster formation from iron and sulfide ions on a scaffold protein, and subsequent delivery to final targets via a series of Fe-S cluster-binding carrier proteins. The process of cluster release from the scaffold/carrier for transfer to the target proteins may be mediated by a dedicated Fe-S cluster chaperone system. In human cells, the chaperones include heat shock protein HSPA9 and the J-type chaperone Hsc20. While the role of chaperones has been somewhat clarified in yeast and bacterial systems, many questions remain over their functional roles in cluster delivery and interactions with a variety of human Fe-S cluster proteins. One such protein, Nfu, has recently been recognized as a potential interaction partner of the chaperone complex. Herein, we examined the ability of human Nfu to function as a carrier by interacting with the human chaperone complex. Human Nfu is shown to bind to both chaperone proteins with binding affinities similar to those observed for IscU binding to the homologous HSPA9 and Hsc20, while Nfu can also stimulate the ATPase activity of HSPA9. Additionally, the chaperone complex was able to promote Nfu function by enhancing the second-order rate constants for Fe-S cluster transfer to target proteins and providing directionality in cluster transfer from Nfu by eliminating promiscuous transfer reactions. Together, these data support a hypothesis in which Nfu can serve as an alternative carrier protein for chaperone-mediated cluster release and delivery in Fe-S cluster biogenesis and trafficking.
Collapse
Affiliation(s)
- Christine Wachnowsky
- Department of Chemistry and Biochemistry, The Ohio State University, Columbus, OH, USA.,The Ohio State Biochemistry Program, The Ohio State University, Columbus, OH, USA
| | - Yushi Liu
- Department of Chemistry and Biochemistry, The Ohio State University, Columbus, OH, USA.,The Ohio State Biochemistry Program, The Ohio State University, Columbus, OH, USA
| | - Taejin Yoon
- Department of Chemistry and Biochemistry, The Ohio State University, Columbus, OH, USA
| | - J A Cowan
- Department of Chemistry and Biochemistry, The Ohio State University, Columbus, OH, USA.,The Ohio State Biochemistry Program, The Ohio State University, Columbus, OH, USA
| |
Collapse
|
17
|
Potts KS, Bowman TV. Modeling Myeloid Malignancies Using Zebrafish. Front Oncol 2017; 7:297. [PMID: 29255698 PMCID: PMC5722844 DOI: 10.3389/fonc.2017.00297] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2017] [Accepted: 11/20/2017] [Indexed: 02/04/2023] Open
Abstract
Human myeloid malignancies represent a substantial disease burden to individuals, with significant morbidity and death. The genetic underpinnings of disease formation and progression remain incompletely understood. Large-scale human population studies have identified a high frequency of potential driver mutations in spliceosomal and epigenetic regulators that contribute to malignancies, such as myelodysplastic syndromes (MDS) and leukemias. The high conservation of cell types and genes between humans and model organisms permits the investigation of the underlying mechanisms of leukemic development and potential therapeutic testing in genetically pliable pre-clinical systems. Due to the many technical advantages, such as large-scale screening, lineage-tracing studies, tumor transplantation, and high-throughput drug screening approaches, zebrafish is emerging as a model system for myeloid malignancies. In this review, we discuss recent advances in MDS and leukemia using the zebrafish model.
Collapse
Affiliation(s)
- Kathryn S Potts
- Department of Developmental and Molecular Biology, Albert Einstein College of Medicine, Bronx, NY, United States.,Gottesman Institute for Stem Cell Biology and Regenerative Medicine, Albert Einstein College of Medicine, Bronx, NY, United States
| | - Teresa V Bowman
- Department of Developmental and Molecular Biology, Albert Einstein College of Medicine, Bronx, NY, United States.,Gottesman Institute for Stem Cell Biology and Regenerative Medicine, Albert Einstein College of Medicine, Bronx, NY, United States.,Department of Medicine (Oncology), Albert Einstein College of Medicine, Bronx, NY, United States
| |
Collapse
|
18
|
Iron metabolism in erythroid cells and patients with congenital sideroblastic anemia. Int J Hematol 2017; 107:44-54. [PMID: 29139060 DOI: 10.1007/s12185-017-2368-0] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2017] [Accepted: 11/08/2017] [Indexed: 01/31/2023]
Abstract
Sideroblastic anemias are anemic disorders characterized by the presence of ring sideroblasts in a patient's bone marrow. These disorders are typically divided into two types, congenital or acquired sideroblastic anemia. Recently, several genes were reported as responsible for congenital sideroblastic anemia; however, the relationship between the function of the gene products and ring sideroblasts is largely unclear. In this review article, we will focus on the iron metabolism in erythroid cells as well as in patients with congenital sideroblastic anemia.
Collapse
|
19
|
Ben-Hamo O, Rosner A, Rabinowitz C, Oren M, Rinkevich B. Coupling astogenic aging in the colonial tunicate Botryllus schlosseri with the stress protein mortalin. Dev Biol 2017; 433:33-46. [PMID: 29128264 DOI: 10.1016/j.ydbio.2017.10.023] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2017] [Revised: 10/30/2017] [Accepted: 10/30/2017] [Indexed: 10/18/2022]
Abstract
Botryllus schlosseri, a colonial marine invertebrate, exhibits three generations of short-lived astogenic modules that continuously grow and die throughout the colony's entire lifespan, within week-long repeating budding cycles (blastogenesis), each consisting of four stages (A-D). At stage D, aging is followed by the complete absorption of adult modules (zooids) via a massive apoptotic process. Here we studied in Botryllus the protein mortalin (HSP70s member), a molecule largely known for its association with aging and proliferation. In-situ hybridization and qPCR assays reveal that mortalin follows the cyclic pattern of blastogenesis. Colonies at blastogenic stage D display the highest mortalin levels, and young modules exhibit elevated mortalin levels compared to old modules. Manipulations of mortalin with the specific allosteric inhibitor MKT-077 has led to a decrease in the modules' growth rate and the development of abnormal somatic/germinal morphologies (primarily in vasculature and in organs such as the endostyle, the stomach and gonads). We therefore propose that mortalin plays a significant role in the astogeny and aging of colonial modules in B. schlosseri, by direct involvement in the regulation of blastogenesis.
Collapse
Affiliation(s)
- Oshrat Ben-Hamo
- Israel Oceanographic and Limnological Research, National Institute of Oceanography, Tel Shikmona, P.O. Box 8030, Haifa 31080, Israel; Department of Marine Biology, Leon H. Charney School of Marine Sciences, University of Haifa, 31905 Haifa, Israel.
| | - Amalia Rosner
- Israel Oceanographic and Limnological Research, National Institute of Oceanography, Tel Shikmona, P.O. Box 8030, Haifa 31080, Israel
| | - Claudette Rabinowitz
- Israel Oceanographic and Limnological Research, National Institute of Oceanography, Tel Shikmona, P.O. Box 8030, Haifa 31080, Israel
| | - Matan Oren
- Israel Oceanographic and Limnological Research, National Institute of Oceanography, Tel Shikmona, P.O. Box 8030, Haifa 31080, Israel; Department of Molecular Biology, Ariel University, Ariel 40700, Israel
| | - Baruch Rinkevich
- Israel Oceanographic and Limnological Research, National Institute of Oceanography, Tel Shikmona, P.O. Box 8030, Haifa 31080, Israel.
| |
Collapse
|
20
|
Liu T, Krysiak K, Shirai CL, Kim S, Shao J, Ndonwi M, Walter MJ. Knockdown of HSPA9 induces TP53-dependent apoptosis in human hematopoietic progenitor cells. PLoS One 2017; 12:e0170470. [PMID: 28178280 PMCID: PMC5298293 DOI: 10.1371/journal.pone.0170470] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2016] [Accepted: 01/05/2017] [Indexed: 11/19/2022] Open
Abstract
Myelodysplastic syndromes (MDS) are the most common adult myeloid blood cancers in the US. Patients have increased apoptosis in their bone marrow cells leading to low peripheral blood counts. The full complement of gene mutations that contribute to increased apoptosis in MDS remains unknown. Up to 25% of MDS patients harbor and acquired interstitial deletion on the long arm of chromosome 5 [del(5q)], creating haploinsufficiency for a large set of genes including HSPA9. Knockdown of HSPA9 in primary human CD34+ hematopoietic progenitor cells significantly inhibits growth and increases apoptosis. We show here that HSPA9 knockdown is associated with increased TP53 expression and activity, resulting in increased expression of target genes BAX and p21. HSPA9 protein interacts with TP53 in CD34+ cells and knockdown of HSPA9 increases nuclear TP53 levels, providing a possible mechanism for regulation of TP53 by HSPA9 haploinsufficiency in hematopoietic cells. Concurrent knockdown of TP53 and HSPA9 rescued the increased apoptosis observed in CD34+ cells following knockdown of HSPA9. Reduction of HSPA9 below 50% results in severe inhibition of cell growth, suggesting that del(5q) cells may be preferentially sensitive to further reductions of HSPA9 below 50%, thus providing a genetic vulnerability to del(5q) cells. Treatment of bone marrow cells with MKT-077, an HSPA9 inhibitor, induced apoptosis in a higher percentage of cells from MDS patients with del(5q) compared to non-del(5q) MDS patients and normal donor cells. Collectively, these findings indicate that reduced levels of HSPA9 may contribute to TP53 activation and increased apoptosis observed in del(5q)-associated MDS.
Collapse
Affiliation(s)
- Tuoen Liu
- Department of Medicine, Division of Oncology, Washington University School of Medicine, St. Louis, Missouri, United States of America
| | - Kilannin Krysiak
- Department of Medicine, Division of Oncology, Washington University School of Medicine, St. Louis, Missouri, United States of America
| | - Cara Lunn Shirai
- Department of Medicine, Division of Oncology, Washington University School of Medicine, St. Louis, Missouri, United States of America
| | - Sanghyun Kim
- Department of Medicine, Division of Oncology, Washington University School of Medicine, St. Louis, Missouri, United States of America
| | - Jin Shao
- Department of Medicine, Division of Oncology, Washington University School of Medicine, St. Louis, Missouri, United States of America
| | - Matthew Ndonwi
- Department of Medicine, Division of Oncology, Washington University School of Medicine, St. Louis, Missouri, United States of America
| | - Matthew J Walter
- Department of Medicine, Division of Oncology, Washington University School of Medicine, St. Louis, Missouri, United States of America
- Siteman Cancer Center, Washington University School of Medicine, St. Louis, Missouri, United States of America
| |
Collapse
|
21
|
Abstract
The zebrafish, Danio rerio, is a well-established, invaluable model system for the study of human cancers. The genetic pathways that drive oncogenesis are highly conserved between zebrafish and humans, and multiple unique attributes of the zebrafish make it a tractable tool for analyzing the underlying cellular processes that give rise to human disease. In particular, the high conservation between human and zebrafish hematopoiesis (Jing & Zon, 2011) has stimulated the development of zebrafish models for human hematopoietic malignancies to elucidate molecular pathogenesis and to expedite the preclinical investigation of novel therapies. While T-cell acute lymphoblastic leukemia was the first transgenic cancer model in zebrafish (Langenau et al., 2003), a wide spectrum of zebrafish models of human hematopoietic malignancies has been established since 2003, largely through transgenesis and genome-editing approaches. This chapter presents key examples that validate the zebrafish as an indispensable model system for the study of hematopoietic malignancies and highlights new models that demonstrate recent advances in the field.
Collapse
Affiliation(s)
- S He
- Harvard Medical School, Boston, MA, United States
| | - C-B Jing
- Harvard Medical School, Boston, MA, United States
| | - A T Look
- Harvard Medical School, Boston, MA, United States
| |
Collapse
|
22
|
Kwan W, North TE. Netting Novel Regulators of Hematopoiesis and Hematologic Malignancies in Zebrafish. Curr Top Dev Biol 2017; 124:125-160. [DOI: 10.1016/bs.ctdb.2016.11.005] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022]
|
23
|
Holowiecki A, O'Shields B, Jenny MJ. Spatiotemporal expression and transcriptional regulation of heme oxygenase and biliverdin reductase genes in zebrafish (Danio rerio) suggest novel roles during early developmental periods of heightened oxidative stress. Comp Biochem Physiol C Toxicol Pharmacol 2017; 191:138-151. [PMID: 27760386 PMCID: PMC5148680 DOI: 10.1016/j.cbpc.2016.10.006] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/07/2016] [Revised: 10/13/2016] [Accepted: 10/13/2016] [Indexed: 02/04/2023]
Abstract
Heme oxygenase 1 (HMOX1) degrades heme into biliverdin, which is subsequently converted to bilirubin by biliverdin reductase (BVRa or BVRb) in a manner analogous to the classic anti-oxidant glutathione-recycling pathway. To gain a better understanding of the potential antioxidant roles the BVR enzymes may play during development, the spatiotemporal expression and transcriptional regulation of zebrafish hmox1a, bvra and bvrb were characterized under basal conditions and in response to pro-oxidant exposure. All three genes displayed spatiotemporal expression patterns consistent with classic hematopoietic progenitors during development. Transient knockdown of Nrf2a did not attenuate the ability to detect bvra or bvrb by ISH, or alter spatial expression patterns in response to cadmium exposure. While hmox1a:mCherry fluorescence was documented within the intermediate cell mass, a transient location of primitive erythrocyte differentiation, expression was not fully attenuated in Nrf2a morphants, but real-time RT-PCR demonstrated a significant reduction in hmox1a expression. Furthermore, Gata-1 knockdown did not attenuate hmox1a:mCherry fluorescence. However, while there was a complete loss of detection of bvrb expression by ISH at 24hpf, bvra expression was greatly attenuated but still detectable in Gata-1 morphants. In contrast, 96 hpf Gata-1 morphants displayed increased bvra and bvrb expression within hematopoietic tissues. Finally, temporal expression patterns of enzymes involved in the generation and maintenance of NADPH were consistent with known changes in the cellular redox state during early zebrafish development. Together, these data suggest that Gata-1 and Nrf2a play differential roles in regulating the heme degradation enzymes during an early developmental period of heightened cellular stress.
Collapse
Affiliation(s)
- Andrew Holowiecki
- Department of Biological Sciences, University of Alabama, Tuscaloosa, AL 35487, USA
| | - Britton O'Shields
- Department of Biological Sciences, University of Alabama, Tuscaloosa, AL 35487, USA
| | - Matthew J Jenny
- Department of Biological Sciences, University of Alabama, Tuscaloosa, AL 35487, USA.
| |
Collapse
|
24
|
Comparison of Integrated Responses to Nonlethal and Lethal Hypothermal Stress in Milkfish (Chanos chanos): A Proteomics Study. PLoS One 2016; 11:e0163538. [PMID: 27657931 PMCID: PMC5033585 DOI: 10.1371/journal.pone.0163538] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2016] [Accepted: 09/10/2016] [Indexed: 11/19/2022] Open
Abstract
Milkfish is an important aquaculture species in Taiwan, and its high mortality during cold snaps in winter usually causes huge economic losses. To understand the effect of hypothermal stress and the corresponding compensatory stress response in milkfish, this study aimed to compare liver and gill protein levels between milkfish exposed to nonlethal (18°C), lethal (16°C), and control (28°C) temperatures. Using a proteomics approach based on two-dimensional electrophoresis and nano-LC-MS/MS analysis, this study identified thirty unique protein spots from milkfish livers and gills for which protein abundance was significantly different between nonlethal, lethal, and control temperature groups. Proteins identified in the liver were classified into three different categories according to their cellular function: (1) anti-oxidative stress, (2) apoptotic pathway, and (3) cytoskeleton. Similarly, proteins identified in the gill were sorted in five different functional categories: (1) cytoskeleton, (2) immune response, (3) protein quality control, (4) energy production, and (5) intracellular homeostasis. Based on functional information derived from the identified proteins, we assumed that different levels of hypothermal stress had a different effect and induced a different cellular response. Upon nonlethal hypothermal stress, the identified proteins were involved in anti-oxidative stress and anti-inflammation pathways, suggesting that milkfish had high levels of oxidative stress in the liver and exhibited inflammation response in the gill. Upon lethal hypothermal stress, however, identified proteins were associated with apoptosis in the liver and regulation of intracellular homeostasis in the gill. The present study provided evidence to illustrate different multi-physiological responses to nonlethal and lethal hypothermal stress in milkfish livers and gills.
Collapse
|
25
|
Tzaneva V, Perry SF. Evidence for a role of heme oxygenase-1 in the control of cardiac function in zebrafish (Danio rerio) larvae exposed to hypoxia. ACTA ACUST UNITED AC 2016; 219:1563-71. [PMID: 26994186 DOI: 10.1242/jeb.136853] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/24/2015] [Accepted: 02/29/2016] [Indexed: 12/15/2022]
Abstract
Carbon monoxide (CO) is a gaseous neurotransmitter produced from the breakdown of heme via heme oxygenase-1 (HO-1; hypoxia-inducible isoform) and heme oxygenase-2 (HO-2; constitutively expressed isoform). In mammals, CO is involved in modulating cardiac function. The role of the HO-1/CO system in the control of heart function in fish, however, is unknown and investigating its physiological function in lower vertebrates will provide a better understanding of the evolution of this regulatory mechanism. We explored the role of the HO-1/CO system in larval zebrafish (Danio rerio) in vivo by investigating the impact of translational gene knockdown of HO-1 on cardiac function. Immunohistochemistry revealed the presence of HO-1 in the pacemaker cells of the heart at 4 days post-fertilization and thus the potential for CO production at these sites. Sham-treated zebrafish larvae (experiencing normal levels of HO-1) significantly increased heart rate (fH) when exposed to hypoxia (PwO2 =30 mmHg). Zebrafish larvae lacking HO-1 expression after morpholino knockdown (morphants) exhibited significantly higher fH under normoxic (but not hypoxic) conditions when compared with sham-treaded fish. The increased fH in HO-1 morphants was rescued (fH was restored to control levels) after treatment of larvae with a CO-releasing molecule (40 µmol l(-1) CORM). The HO-1-deficient larvae developed significantly larger ventricles and when exposed to hypoxia they displayed higher cardiac output ([Formula: see text]) and stroke volume (SV). These results suggest that under hypoxic conditions, HO-1 regulates [Formula: see text] and SV presumably via the production of CO. Overall, this study provides a better understanding of the role of the HO-1/CO system in controlling heart function in lower vertebrates. We demonstrate for the first time the ability for CO to be produced in presumptive pacemaker cells of the heart where it plays an inhibitory role in setting the resting cardiac frequency.
Collapse
Affiliation(s)
- Velislava Tzaneva
- Department of Biology, University of Ottawa, 30 Marie Curie, Ottawa, ON, Canada K1N 6N5
| | - Steve F Perry
- Department of Biology, University of Ottawa, 30 Marie Curie, Ottawa, ON, Canada K1N 6N5
| |
Collapse
|
26
|
Wan Y, Zhang Q, Zhang Z, Song B, Wang X, Zhang Y, Jia Q, Cheng T, Zhu X, Leung AYH, Yuan W, Jia H, Fang X. Transcriptome analysis reveals a ribosome constituents disorder involved in the RPL5 downregulated zebrafish model of Diamond-Blackfan anemia. BMC Med Genomics 2016; 9:13. [PMID: 26961822 PMCID: PMC4785739 DOI: 10.1186/s12920-016-0174-9] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2015] [Accepted: 03/03/2016] [Indexed: 01/06/2023] Open
Abstract
BACKGROUND Diamond-Blackfan anemia (DBA) was the first ribosomopathy associated with mutations in ribosome protein (RP) genes. The clinical phenotypes of DBA include failure of erythropoiesis, congenital anomalies and cancer predisposition. Mutations in RPL5 are reported in approximately 9 ~ 21 % of DBA patients, which represents the most common pathological condition related to a large-subunit ribosomal protein. However, it remains unclear how RPL5 downregulation results in severe phenotypes of this disease. RESULTS In this study, we generated a zebrafish model of DBA with RPL5 morphants and implemented high-throughput RNA-seq and ncRNA-seq to identify key genes, lncRNAs, and miRNAs during zebrafish development and hematopoiesis. We demonstrated that RPL5 is required for both primitive and definitive hematopoiesis processes. By comparing with other DBA zebrafish models and processing functional coupling network, we identified some common regulated genes, lncRNAs and miRNAs, that might play important roles in development and hematopoiesis. CONCLUSIONS Ribosome biogenesis and translation process were affected more in RPL5 MO than in other RP MOs. Both P53 dependent (for example, cell cycle pathway) and independent pathways (such as Aminoacyl-tRNA biosynthesis pathway) play important roles in DBA pathology. Our results therefore provide a comprehensive basis for the study of molecular pathogenesis of RPL5-mediated DBA and other ribosomopathies.
Collapse
Affiliation(s)
- Yang Wan
- />State Key Laboratory of Experimental Hematology, Institute of Hematology and Blood Diseases Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Tianjin, 300020 China
| | - Qian Zhang
- />CAS Key Laboratory of Genome Sciences and Information, Beijing Institute of Genomics, Chinese Academy of Sciences, Beijing, 100101 China
| | - Zhaojun Zhang
- />CAS Key Laboratory of Genome Sciences and Information, Beijing Institute of Genomics, Chinese Academy of Sciences, Beijing, 100101 China
| | - Binfeng Song
- />Key Laboratory of Molecular Biophysics of Ministry of Education, College of Life Science and Technology, Center for Human Genome Research, Huazhong University of Science and Technology, Wuhan, Hubei 430074 China
| | - Xiaomin Wang
- />State Key Laboratory of Experimental Hematology, Institute of Hematology and Blood Diseases Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Tianjin, 300020 China
| | - Yingchi Zhang
- />State Key Laboratory of Experimental Hematology, Institute of Hematology and Blood Diseases Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Tianjin, 300020 China
| | - Qiong Jia
- />Key Laboratory of Molecular Biophysics of Ministry of Education, College of Life Science and Technology, Center for Human Genome Research, Huazhong University of Science and Technology, Wuhan, Hubei 430074 China
| | - Tao Cheng
- />State Key Laboratory of Experimental Hematology, Institute of Hematology and Blood Diseases Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Tianjin, 300020 China
| | - Xiaofan Zhu
- />State Key Laboratory of Experimental Hematology, Institute of Hematology and Blood Diseases Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Tianjin, 300020 China
| | | | - Weiping Yuan
- />State Key Laboratory of Experimental Hematology, Institute of Hematology and Blood Diseases Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Tianjin, 300020 China
| | - Haibo Jia
- />Key Laboratory of Molecular Biophysics of Ministry of Education, College of Life Science and Technology, Center for Human Genome Research, Huazhong University of Science and Technology, Wuhan, Hubei 430074 China
| | - Xiangdong Fang
- />CAS Key Laboratory of Genome Sciences and Information, Beijing Institute of Genomics, Chinese Academy of Sciences, Beijing, 100101 China
| |
Collapse
|
27
|
Bellipanni G, Cappello F, Scalia F, Conway de Macario E, Macario AJ, Giordano A. Zebrafish as a Model for the Study of Chaperonopathies. J Cell Physiol 2016; 231:2107-14. [DOI: 10.1002/jcp.25319] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2016] [Accepted: 01/25/2016] [Indexed: 11/08/2022]
Affiliation(s)
- Gianfranco Bellipanni
- Sbarro Institute for Cancer Research and Molecular Medicine; Philadelphia Pennsylvania
- Department of Biology; College of Science and Technology, Temple University; Philadelphia Pennsylvania
- Euro-Mediterranean Institute of Science and Technology (IEMEST); Palermo Italy
| | - Francesco Cappello
- Department of Biology; College of Science and Technology, Temple University; Philadelphia Pennsylvania
- Euro-Mediterranean Institute of Science and Technology (IEMEST); Palermo Italy
- Department of Experimental Biomedicine and Clinical Neuroscience; University of Palermo; Palermo Italy
| | - Federica Scalia
- Department of Experimental Biomedicine and Clinical Neuroscience; University of Palermo; Palermo Italy
| | - Everly Conway de Macario
- Department of Microbiology and Immunology; School of Medicine, University of Maryland at Baltimore and IMET; Baltimore Maryland
| | - Alberto J.L. Macario
- Euro-Mediterranean Institute of Science and Technology (IEMEST); Palermo Italy
- Department of Microbiology and Immunology; School of Medicine, University of Maryland at Baltimore and IMET; Baltimore Maryland
| | - Antonio Giordano
- Sbarro Institute for Cancer Research and Molecular Medicine; Philadelphia Pennsylvania
- Department of Biology; College of Science and Technology, Temple University; Philadelphia Pennsylvania
| |
Collapse
|
28
|
Shan Y, Cortopassi G. Mitochondrial Hspa9/Mortalin regulates erythroid differentiation via iron-sulfur cluster assembly. Mitochondrion 2015; 26:94-103. [PMID: 26702583 DOI: 10.1016/j.mito.2015.12.005] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2015] [Revised: 09/04/2015] [Accepted: 12/11/2015] [Indexed: 01/30/2023]
Abstract
Mitochondrial iron-sulfur cluster (ISC) biogenesis provides iron-sulfur cofactors to several mitochondrial proteins, but the extent to which ISC biogenesis regulates hematopoiesis has been unclear. The blood disease Myelodysplastic syndrome (MDS) is characterized by ineffective hematopoiesis, and the disease overlaps with the gene Hspa9/Mortalin in multiple ways: the HSPA9 locus maps to 5q31.2 that is frequently deleted in human MDS; mutant Hspa9 causes zebrafish MDS; and Hspa9 knockdown mice have decreased hematopoiesis. We show here that HSPA9 functions in mitochondrial ISC biogenesis, and is required for erythroid differentiation. HSPA9 interacts with and stabilizes the mitochondrial ISC biogenesis proteins frataxin, Nfs1, ISCU, and Nfu. MDS-causing mutations in HSPA9 protein change its interactions with ISC biogenesis proteins. Depletion of HSPA9 decreases aconitase activity, which requires an ISC at its active site, but not that of the non-ISC requiring malate dehydrogenase, and increases IRP1 binding activity. In erythroid cell lines, Hspa9 depletion inhibited erythroid differentiation, post-transcriptionally regulating the expression of Alas2 and FeCH, as expected through known ISC control of the IRE response elements in these genes. By contrast, the Alas2 open reading frame rescued the Hspa9-dependent defect in erythroid differentiation, but not when uncoupled from its 5'-IRE sequence. Thus, Hspa9 depletion causes a mitochondrial ISC deficit, altering IRP1-IRE binding and FeCH stability, which consequently inhibits Alas2 translation, heme synthesis, and erythroid differentiation, i.e.: Hspa9->ISC->IRP/IRE->Alas2->heme synthesis->erythroid differentiation. Thus Hspa9 regulates erythroid differentiation through ISC cluster assembly, providing a pathophysiological mechanism for an MDS subtype characterized by HSPA9 haploinsufficiency, and suggests hemin and other pharmacological stimulators of ISC synthesis as potential routes to therapy.
Collapse
Affiliation(s)
- Yuxi Shan
- University of California, Davis, VM: Dept. of Molecular Biosciences, One Shields Ave., Davis 95616, USA.
| | - Gino Cortopassi
- University of California, Davis, VM: Dept. of Molecular Biosciences, One Shields Ave., Davis 95616, USA.
| |
Collapse
|
29
|
Royer-Bertrand B, Castillo-Taucher S, Moreno-Salinas R, Cho TJ, Chae JH, Choi M, Kim OH, Dikoglu E, Campos-Xavier B, Girardi E, Superti-Furga G, Bonafé L, Rivolta C, Unger S, Superti-Furga A. Mutations in the heat-shock protein A9 (HSPA9) gene cause the EVEN-PLUS syndrome of congenital malformations and skeletal dysplasia. Sci Rep 2015; 5:17154. [PMID: 26598328 PMCID: PMC4657157 DOI: 10.1038/srep17154] [Citation(s) in RCA: 53] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2015] [Accepted: 10/26/2015] [Indexed: 11/18/2022] Open
Abstract
We and others have reported mutations in LONP1, a gene coding for a mitochondrial chaperone and protease, as the cause of the human CODAS (cerebral, ocular, dental, auricular and skeletal) syndrome (MIM 600373). Here, we delineate a similar but distinct condition that shares the epiphyseal, vertebral and ocular changes of CODAS but also included severe microtia, nasal hypoplasia, and other malformations, and for which we propose the name of EVEN-PLUS syndrome for epiphyseal, vertebral, ear, nose, plus associated findings. In three individuals from two families, no mutation in LONP1 was found; instead, we found biallelic mutations in HSPA9, the gene that codes for mHSP70/mortalin, another highly conserved mitochondrial chaperone protein essential in mitochondrial protein import, folding, and degradation. The functional relationship between LONP1 and HSPA9 in mitochondrial protein chaperoning and the overlapping phenotypes of CODAS and EVEN-PLUS delineate a family of “mitochondrial chaperonopathies” and point to an unexplored role of mitochondrial chaperones in human embryonic morphogenesis.
Collapse
Affiliation(s)
- Beryl Royer-Bertrand
- Department of Medical Genetics, University of Lausanne, Lausanne, Switzerland.,Centre for Molecular Diseases, Department of Pediatrics, Lausanne University Hospital (CHUV), Lausanne, Switzerland
| | - Silvia Castillo-Taucher
- Sección Genética, Hospital Clínico Universidad de Chile, and Sección Citogenética, Laboratorio, Clínica Alemana de Santiago, Santiago, Chile
| | - Rodrigo Moreno-Salinas
- Unidad de Genética, Hospital Regional Rancagua, Rancagua, Chile; and ICBM, Facultad de Medicina, Universidad de Chile, Santiago, Chile
| | - Tae-Joon Cho
- Division of Pediatric Orthopaedics, Seoul National University Children's Hospital, Seoul, Republic of Korea
| | - Jong-Hee Chae
- Department of Pediatrics, Pediatric Clinical Neuroscience Center, Seoul National University Children's Hospital, Seoul, Republic of Korea
| | - Murim Choi
- Department of Biomedical Sciences, Seoul National University College of Medicine, Seoul, Republic of Korea
| | - Ok-Hwa Kim
- Department of Radiology, Woorisoa Children's Hospital, Saemalro, Guro-gu, Seoul 08291, Republic of Korea
| | - Esra Dikoglu
- Centre for Molecular Diseases, Department of Pediatrics, Lausanne University Hospital (CHUV), Lausanne, Switzerland
| | - Belinda Campos-Xavier
- Centre for Molecular Diseases, Department of Pediatrics, Lausanne University Hospital (CHUV), Lausanne, Switzerland
| | - Enrico Girardi
- CeMM Research Center for Molecular Medicine of the Austrian Academy of Sciences, 1090 Vienna, Austria
| | - Giulio Superti-Furga
- CeMM Research Center for Molecular Medicine of the Austrian Academy of Sciences, 1090 Vienna, Austria
| | - Luisa Bonafé
- Centre for Molecular Diseases, Department of Pediatrics, Lausanne University Hospital (CHUV), Lausanne, Switzerland
| | - Carlo Rivolta
- Department of Medical Genetics, University of Lausanne, Lausanne, Switzerland
| | - Sheila Unger
- Medical Genetics Service, Lausanne University Hospital (CHUV) and University of Lausanne, Lausanne, Switzerland
| | - Andrea Superti-Furga
- Centre for Molecular Diseases, Department of Pediatrics, Lausanne University Hospital (CHUV), Lausanne, Switzerland.,Department of Pediatrics and Pediatric Surgery, University of Lausanne and Lausanne University Hospital (CHUV), Lausanne, Switzerland
| |
Collapse
|
30
|
Congenital sideroblastic anemia due to mutations in the mitochondrial HSP70 homologue HSPA9. Blood 2015; 126:2734-8. [PMID: 26491070 DOI: 10.1182/blood-2015-09-659854] [Citation(s) in RCA: 52] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2015] [Accepted: 10/01/2015] [Indexed: 12/19/2022] Open
Abstract
The congenital sideroblastic anemias (CSAs) are relatively uncommon diseases characterized by defects in mitochondrial heme synthesis, iron-sulfur (Fe-S) cluster biogenesis, or protein synthesis. Here we demonstrate that mutations in HSPA9, a mitochondrial HSP70 homolog located in the chromosome 5q deletion syndrome 5q33 critical deletion interval and involved in mitochondrial Fe-S biogenesis, result in CSA inherited as an autosomal recessive trait. In a fraction of patients with just 1 severe loss-of-function allele, expression of the clinical phenotype is associated with a common coding single nucleotide polymorphism in trans that correlates with reduced messenger RNA expression and results in a pseudodominant pattern of inheritance.
Collapse
|
31
|
HSP90 and HSP70: Implication in Inflammation Processes and Therapeutic Approaches for Myeloproliferative Neoplasms. Mediators Inflamm 2015; 2015:970242. [PMID: 26549943 PMCID: PMC4624912 DOI: 10.1155/2015/970242] [Citation(s) in RCA: 58] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2015] [Accepted: 09/27/2015] [Indexed: 12/23/2022] Open
Abstract
Myeloproliferative neoplasms (MPN) are clonal stem cell disorders that lead to the excessive production of one or more blood cell lineages. It has been reported that, in most MPN, inflammatory cytokines are frequently increased, indicating that inflammation plays a crucial role in these disorders. Heat shock proteins (HSP) are induced in response to many stressful conditions from heat shock to hypoxia and inflammation. Besides their chaperone and cytoprotective functions, HSPs are key players during inflammation, hence the term “chaperokine.” Through their chaperone activity, HSP90, a stabilizer of many oncogenes (e.g., JAK2), and HSP70, a powerful antiapoptotic chaperone, tightly regulate Nuclear Factor-kappa B signalling, a critical pathway in mediating inflammatory responses. In light of this potential, several HSP90 inhibitors have been generated as anticancer agents able to degrade oncogenes. As it turns out, however, these drugs are also potent inhibitors of the inflammatory response in various diseases. Given the chaperone potential of HSP70 and the fact that HSP90 inhibitors induce HSP70, interest in HSP70 inhibitors is also increasing. Here, we focus on the implication of HSP90 and HSP70 in inflammatory responses and on the emergence of new therapeutic approaches in MPN based on HSP inhibitors.
Collapse
|
32
|
Rissone A, Weinacht KG, la Marca G, Bishop K, Giocaliere E, Jagadeesh J, Felgentreff K, Dobbs K, Al-Herz W, Jones M, Chandrasekharappa S, Kirby M, Wincovitch S, Simon KL, Itan Y, DeVine A, Schlaeger T, Schambach A, Sood R, Notarangelo LD, Candotti F. Reticular dysgenesis-associated AK2 protects hematopoietic stem and progenitor cell development from oxidative stress. ACTA ACUST UNITED AC 2015; 212:1185-202. [PMID: 26150473 PMCID: PMC4516804 DOI: 10.1084/jem.20141286] [Citation(s) in RCA: 42] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2014] [Accepted: 06/01/2015] [Indexed: 12/23/2022]
Abstract
Rissone et al. demonstrate that adenylate kinase AK2, an enzyme mutated in reticular dysgenesis (RD) in humans, prevents oxidative stress during hematopoiesis. Using a zebrafish model, as well as induced pluripotent stem cells derived from an RD patient, they find that AK2 deficiency affects hematopoietic stem and progenitor development with increased oxidative stress. Antioxidant treatment rescues the hematopoietic phenotypes. Adenylate kinases (AKs) are phosphotransferases that regulate the cellular adenine nucleotide composition and play a critical role in the energy homeostasis of all tissues. The AK2 isoenzyme is expressed in the mitochondrial intermembrane space and is mutated in reticular dysgenesis (RD), a rare form of severe combined immunodeficiency (SCID) in humans. RD is characterized by a maturation arrest in the myeloid and lymphoid lineages, leading to early onset, recurrent, and overwhelming infections. To gain insight into the pathophysiology of RD, we studied the effects of AK2 deficiency using the zebrafish model and induced pluripotent stem cells (iPSCs) derived from fibroblasts of an RD patient. In zebrafish, Ak2 deficiency affected hematopoietic stem and progenitor cell (HSPC) development with increased oxidative stress and apoptosis. AK2-deficient iPSCs recapitulated the characteristic myeloid maturation arrest at the promyelocyte stage and demonstrated an increased AMP/ADP ratio, indicative of an energy-depleted adenine nucleotide profile. Antioxidant treatment rescued the hematopoietic phenotypes in vivo in ak2 mutant zebrafish and restored differentiation of AK2-deficient iPSCs into mature granulocytes. Our results link hematopoietic cell fate in AK2 deficiency to cellular energy depletion and increased oxidative stress. This points to the potential use of antioxidants as a supportive therapeutic modality for patients with RD.
Collapse
Affiliation(s)
- Alberto Rissone
- Disorders of Immunity Section, Genetics and Molecular Biology Branch; Zebrafish Core and Oncogenesis and Development Section, Translational and Functional Genomics Branch; Genomics Core, Cancer Genetics and Comparative Genomics Branch; Division of Intramural Research Flow Cytometry Core; and Cytogenetics and Microscopy Core, Genetic Disease Research Branch; National Human Genome Research Institute, National Institutes of Health, Bethesda, MD 20892
| | - Katja Gabriele Weinacht
- Division of Hematology/Oncology and Division of Immunology, Boston Children's Hospital, Boston, MA 02115 Department of Pediatric Oncology, Dana-Farber Cancer Institute, Boston, MA 02115
| | - Giancarlo la Marca
- Department of Neurosciences, Psychology, Pharmacology, and Child Health, University of Florence, 51039 Florence, Italy Meyer Children's University Hospital, 50141 Florence, Italy
| | - Kevin Bishop
- Disorders of Immunity Section, Genetics and Molecular Biology Branch; Zebrafish Core and Oncogenesis and Development Section, Translational and Functional Genomics Branch; Genomics Core, Cancer Genetics and Comparative Genomics Branch; Division of Intramural Research Flow Cytometry Core; and Cytogenetics and Microscopy Core, Genetic Disease Research Branch; National Human Genome Research Institute, National Institutes of Health, Bethesda, MD 20892
| | | | - Jayashree Jagadeesh
- Disorders of Immunity Section, Genetics and Molecular Biology Branch; Zebrafish Core and Oncogenesis and Development Section, Translational and Functional Genomics Branch; Genomics Core, Cancer Genetics and Comparative Genomics Branch; Division of Intramural Research Flow Cytometry Core; and Cytogenetics and Microscopy Core, Genetic Disease Research Branch; National Human Genome Research Institute, National Institutes of Health, Bethesda, MD 20892
| | - Kerstin Felgentreff
- Division of Hematology/Oncology and Division of Immunology, Boston Children's Hospital, Boston, MA 02115
| | - Kerry Dobbs
- Division of Hematology/Oncology and Division of Immunology, Boston Children's Hospital, Boston, MA 02115
| | - Waleed Al-Herz
- Department of Pediatrics, Faculty of Medicine, Kuwait University, 13110 Kuwait City, Kuwait Allergy and Clinical Immunology Unit, Pediatric Department, Al-Sabah Hospital, 70459 Kuwait City, Kuwait
| | - Marypat Jones
- Disorders of Immunity Section, Genetics and Molecular Biology Branch; Zebrafish Core and Oncogenesis and Development Section, Translational and Functional Genomics Branch; Genomics Core, Cancer Genetics and Comparative Genomics Branch; Division of Intramural Research Flow Cytometry Core; and Cytogenetics and Microscopy Core, Genetic Disease Research Branch; National Human Genome Research Institute, National Institutes of Health, Bethesda, MD 20892
| | - Settara Chandrasekharappa
- Disorders of Immunity Section, Genetics and Molecular Biology Branch; Zebrafish Core and Oncogenesis and Development Section, Translational and Functional Genomics Branch; Genomics Core, Cancer Genetics and Comparative Genomics Branch; Division of Intramural Research Flow Cytometry Core; and Cytogenetics and Microscopy Core, Genetic Disease Research Branch; National Human Genome Research Institute, National Institutes of Health, Bethesda, MD 20892
| | - Martha Kirby
- Disorders of Immunity Section, Genetics and Molecular Biology Branch; Zebrafish Core and Oncogenesis and Development Section, Translational and Functional Genomics Branch; Genomics Core, Cancer Genetics and Comparative Genomics Branch; Division of Intramural Research Flow Cytometry Core; and Cytogenetics and Microscopy Core, Genetic Disease Research Branch; National Human Genome Research Institute, National Institutes of Health, Bethesda, MD 20892
| | - Stephen Wincovitch
- Disorders of Immunity Section, Genetics and Molecular Biology Branch; Zebrafish Core and Oncogenesis and Development Section, Translational and Functional Genomics Branch; Genomics Core, Cancer Genetics and Comparative Genomics Branch; Division of Intramural Research Flow Cytometry Core; and Cytogenetics and Microscopy Core, Genetic Disease Research Branch; National Human Genome Research Institute, National Institutes of Health, Bethesda, MD 20892
| | - Karen Lyn Simon
- Disorders of Immunity Section, Genetics and Molecular Biology Branch; Zebrafish Core and Oncogenesis and Development Section, Translational and Functional Genomics Branch; Genomics Core, Cancer Genetics and Comparative Genomics Branch; Division of Intramural Research Flow Cytometry Core; and Cytogenetics and Microscopy Core, Genetic Disease Research Branch; National Human Genome Research Institute, National Institutes of Health, Bethesda, MD 20892
| | - Yuval Itan
- St. Giles Laboratory of Human Genetics of Infectious Disease, Rockefeller Branch, The Rockefeller University, New York, NY 10065
| | - Alex DeVine
- Division of Hematology/Oncology and Division of Immunology, Boston Children's Hospital, Boston, MA 02115
| | - Thorsten Schlaeger
- Division of Hematology/Oncology and Division of Immunology, Boston Children's Hospital, Boston, MA 02115
| | - Axel Schambach
- Division of Hematology/Oncology and Division of Immunology, Boston Children's Hospital, Boston, MA 02115 Institute of Experimental Hematology, Hannover Medical School, 30625 Hannover, Germany
| | - Raman Sood
- Disorders of Immunity Section, Genetics and Molecular Biology Branch; Zebrafish Core and Oncogenesis and Development Section, Translational and Functional Genomics Branch; Genomics Core, Cancer Genetics and Comparative Genomics Branch; Division of Intramural Research Flow Cytometry Core; and Cytogenetics and Microscopy Core, Genetic Disease Research Branch; National Human Genome Research Institute, National Institutes of Health, Bethesda, MD 20892 Disorders of Immunity Section, Genetics and Molecular Biology Branch; Zebrafish Core and Oncogenesis and Development Section, Translational and Functional Genomics Branch; Genomics Core, Cancer Genetics and Comparative Genomics Branch; Division of Intramural Research Flow Cytometry Core; and Cytogenetics and Microscopy Core, Genetic Disease Research Branch; National Human Genome Research Institute, National Institutes of Health, Bethesda, MD 20892
| | - Luigi D Notarangelo
- Division of Hematology/Oncology and Division of Immunology, Boston Children's Hospital, Boston, MA 02115 Harvard Stem Cell Institute, Harvard University, Cambridge, MA 02138
| | - Fabio Candotti
- Disorders of Immunity Section, Genetics and Molecular Biology Branch; Zebrafish Core and Oncogenesis and Development Section, Translational and Functional Genomics Branch; Genomics Core, Cancer Genetics and Comparative Genomics Branch; Division of Intramural Research Flow Cytometry Core; and Cytogenetics and Microscopy Core, Genetic Disease Research Branch; National Human Genome Research Institute, National Institutes of Health, Bethesda, MD 20892 Division of Immunology and Allergy, University Hospital of Lausanne, 1011 Lausanne, Switzerland
| |
Collapse
|
33
|
Krysiak K, Tibbitts JF, Shao J, Liu T, Ndonwi M, Walter MJ. Reduced levels of Hspa9 attenuate Stat5 activation in mouse B cells. Exp Hematol 2015; 43:319-30.e10. [PMID: 25550197 PMCID: PMC4375022 DOI: 10.1016/j.exphem.2014.12.005] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2014] [Revised: 12/16/2014] [Accepted: 12/18/2014] [Indexed: 11/23/2022]
Abstract
HSPA9 is located on chromosome 5q31.2 in humans, a region that is commonly deleted in patients with myeloid malignancies [del(5q)], including myelodysplastic syndrome (MDS). HSPA9 expression is reduced by 50% in patients with del(5q)-associated MDS, consistent with haploinsufficient levels. Zebrafish mutants and knockdown studies in human and mouse cells have implicated a role for HSPA9 in hematopoiesis. To comprehensively evaluate the effects of Hspa9 haploinsufficiency on hematopoiesis, we generated an Hspa9 knockout mouse model. Although homozygous knockout of Hspa9 is embryonically lethal, mice with heterozygous deletion of Hspa9 (Hspa9(+/-)) are viable and have a 50% reduction in Hspa9 expression. Hspa9(+/-) mice have normal basal hematopoiesis and do not develop MDS. However, Hspa9(+/-) mice have a cell-intrinsic reduction in bone marrow colony-forming unit-PreB colony formation without alterations in the number of B-cell progenitors in vivo, consistent with a functional defect in Hspa9(+/-) B-cell progenitors. We further reduced Hspa9 expression (<50%) using RNA interference and observed reduced B-cell progenitors in vivo, indicating that appropriate levels (≥50%) of Hspa9 are required for normal B lymphopoiesis in vivo. Knockdown of Hspa9 in an interleukin 7 (IL-7)-dependent mouse B-cell line reduced signal transducer and activator of transcription 5 (Stat5) phosphorylation following IL-7 receptor stimulation, supporting a role for Hspa9 in Stat5 signaling in B cells. Collectively, these data imply a role for Hspa9 in B lymphopoiesis and Stat5 activation downstream of IL-7 signaling.
Collapse
Affiliation(s)
- Kilannin Krysiak
- Department of Medicine, Washington University School of Medicine, St. Louis, MO, USA; Division of Oncology, Washington University School of Medicine, St. Louis, MO, USA
| | - Justin F Tibbitts
- Department of Medicine, Washington University School of Medicine, St. Louis, MO, USA; Division of Oncology, Washington University School of Medicine, St. Louis, MO, USA
| | - Jin Shao
- Department of Medicine, Washington University School of Medicine, St. Louis, MO, USA; Division of Oncology, Washington University School of Medicine, St. Louis, MO, USA
| | - Tuoen Liu
- Department of Medicine, Washington University School of Medicine, St. Louis, MO, USA; Division of Oncology, Washington University School of Medicine, St. Louis, MO, USA
| | - Matthew Ndonwi
- Department of Medicine, Washington University School of Medicine, St. Louis, MO, USA; Division of Oncology, Washington University School of Medicine, St. Louis, MO, USA
| | - Matthew J Walter
- Department of Medicine, Washington University School of Medicine, St. Louis, MO, USA; Division of Oncology, Washington University School of Medicine, St. Louis, MO, USA; Department of Genetics, Washington University School of Medicine, St. Louis, MO, USA; Siteman Cancer Center, Washington University School of Medicine, St. Louis, MO, USA.
| |
Collapse
|
34
|
Zebrafish as a model for leukemia and other hematopoietic disorders. J Hematol Oncol 2015; 8:29. [PMID: 25884214 PMCID: PMC4389495 DOI: 10.1186/s13045-015-0126-4] [Citation(s) in RCA: 44] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2015] [Accepted: 03/11/2015] [Indexed: 01/24/2023] Open
Abstract
Zebrafish is an established model for the study of vertebrate development, and is especially amenable for investigating hematopoiesis, where there is strong conservation of key lineages, genes, and developmental processes with humans. Over recent years, zebrafish has been increasingly utilized as a model for a range of human hematopoietic diseases, including malignancies. This review provides an overview of zebrafish hematopoiesis and describes its application as a model of leukemia and other hematopoietic disorders.
Collapse
|
35
|
Bermejo-Nogales A, Nederlof M, Benedito-Palos L, Ballester-Lozano GF, Folkedal O, Olsen RE, Sitjà-Bobadilla A, Pérez-Sánchez J. Metabolic and transcriptional responses of gilthead sea bream (Sparus aurata L.) to environmental stress: new insights in fish mitochondrial phenotyping. Gen Comp Endocrinol 2014; 205:305-15. [PMID: 24792819 DOI: 10.1016/j.ygcen.2014.04.016] [Citation(s) in RCA: 45] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/11/2013] [Revised: 04/09/2014] [Accepted: 04/13/2014] [Indexed: 12/24/2022]
Abstract
The aim of the current study was to phenotype fish metabolism and the transcriptionally-mediated response of hepatic mitochondria of gilthead sea bream to intermittent and repetitive environmental stressors: (i) changes in water temperature (T-ST), (ii) changes in water level and chasing (C-ST) and (iii) multiple sensory perception stressors (M-ST). Gene expression profiling was done using a quantitative PCR array of 60 mitochondria-related genes, selected as markers of transcriptional regulation, oxidative metabolism, respiration uncoupling, antioxidant defense, protein import/folding/assembly, and mitochondrial dynamics and apoptosis. The mitochondrial phenotype mirrored changes in fish performance, haematology and lactate production. T-ST especially up-regulated transcriptional factors (PGC1α, NRF1, NRF2), rate limiting enzymes of fatty acid β-oxidation (CPT1A) and tricarboxylic acid cycle (CS), membrane translocases (Tim/TOM complex) and molecular chaperones (mtHsp10, mtHsp60, mtHsp70) to improve the oxidative capacity in a milieu of a reduced feed intake and impaired haematology. The lack of mitochondrial response, increased production of lactate and negligible effects on growth performance in C-ST fish were mostly considered as a switch from aerobic to anaerobic metabolism. A strong down-regulation of PGC1α, NRF1, NRF2, CPT1A, CS and markers of mitochondrial dynamics and apoptosis (BAX, BCLX, MFN2, MIRO2) occurred in M-ST fish in association with the greatest circulating cortisol concentration and a reduced lactate production and feed efficiency, which represents a metabolic condition with the highest allostatic load score. These findings evidence a high mitochondrial plasticity against stress stimuli, providing new insights to define the threshold level of stress condition in fish.
Collapse
Affiliation(s)
- Azucena Bermejo-Nogales
- Nutrigenomics and Fish Growth Endocrinology Group, Department of Marine Species Biology, Culture and Pathology, Institute of Aquaculture Torre de la Sal, IATS-CSIC, 12595 Ribera de Cabanes s/n, Castellón, Spain.
| | - Marit Nederlof
- Aquaculture and Fisheries Group, Wageningen University, De Elst, 6708 WD Wageningen, The Netherlands.
| | - Laura Benedito-Palos
- Nutrigenomics and Fish Growth Endocrinology Group, Department of Marine Species Biology, Culture and Pathology, Institute of Aquaculture Torre de la Sal, IATS-CSIC, 12595 Ribera de Cabanes s/n, Castellón, Spain.
| | - Gabriel F Ballester-Lozano
- Nutrigenomics and Fish Growth Endocrinology Group, Department of Marine Species Biology, Culture and Pathology, Institute of Aquaculture Torre de la Sal, IATS-CSIC, 12595 Ribera de Cabanes s/n, Castellón, Spain.
| | - Ole Folkedal
- Institute of Marine Research Matre, 5984 Matredal, Norway.
| | | | - Ariadna Sitjà-Bobadilla
- Fish Pathology Group, Department of Marine Species Biology, Culture and Pathology, Institute of Aquaculture Torre de la Sal, IATS-CSIC, 12595 Ribera de Cabanes s/n, Castellón, Spain.
| | - Jaume Pérez-Sánchez
- Nutrigenomics and Fish Growth Endocrinology Group, Department of Marine Species Biology, Culture and Pathology, Institute of Aquaculture Torre de la Sal, IATS-CSIC, 12595 Ribera de Cabanes s/n, Castellón, Spain.
| |
Collapse
|
36
|
Samaddar M, Goswami AV, Purushotham J, Hegde P, D'Silva P. Role of the loop L4,5 in allosteric regulation in mtHsp70s: in vivo significance of domain communication and its implications in protein translocation. Mol Biol Cell 2014; 25:2129-42. [PMID: 24829379 PMCID: PMC4091826 DOI: 10.1091/mbc.e14-03-0821] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/23/2023] Open
Abstract
The SBD loop L4,5 in mtHsp70s functions synergistically with the linker region to maintain the interdomain interface governing protein translocation and mitochondrial biogenesis. Intragenic suppressors of a communication-impaired L4,5 mutant reveal molecular insights into the allosteric regulation of mtHsp70s at the in vivo level. Mitochondrial Hsp70 (mtHsp70) is essential for a vast repertoire of functions, including protein import, and requires effective interdomain communication for efficient partner-protein interactions. However, the in vivo functional significance of allosteric regulation in eukaryotes is poorly defined. Using integrated biochemical and yeast genetic approaches, we provide compelling evidence that a conserved substrate-binding domain (SBD) loop, L4,5, plays a critical role in allosteric communication governing mtHsp70 chaperone functions across species. In yeast, a temperature-sensitive L4,5 mutation (E467A) disrupts bidirectional domain communication, leading to compromised protein import and mitochondrial function. Loop L4,5 functions synergistically with the linker in modulating the allosteric interface and conformational transitions between SBD and the nucleotide-binding domain (NBD), thus regulating interdomain communication. Second-site intragenic suppressors of E467A isolated within the SBD suppress domain communication defects by conformationally altering the allosteric interface, thereby restoring import and growth phenotypes. Strikingly, the suppressor mutations highlight that restoration of communication from NBD to SBD alone is the minimum essential requirement for effective in vivo function when primed at higher basal ATPase activity, mimicking the J-protein–bound state. Together these findings provide the first mechanistic insights into critical regions within the SBD of mtHsp70s regulating interdomain communication, thus highlighting its importance in protein translocation and mitochondrial biogenesis.
Collapse
Affiliation(s)
- Madhuja Samaddar
- Department of Biochemistry, Indian Institute of Science, Bangalore 560012, India
| | | | - Jaya Purushotham
- Department of Biochemistry, Indian Institute of Science, Bangalore 560012, India
| | - Pushpa Hegde
- Department of Biochemistry, Indian Institute of Science, Bangalore 560012, India
| | - Patrick D'Silva
- Department of Biochemistry, Indian Institute of Science, Bangalore 560012, India
| |
Collapse
|
37
|
Steele SL, Prykhozhij SV, Berman JN. Zebrafish as a model system for mitochondrial biology and diseases. Transl Res 2014; 163:79-98. [PMID: 24055494 DOI: 10.1016/j.trsl.2013.08.008] [Citation(s) in RCA: 40] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/24/2013] [Revised: 08/21/2013] [Accepted: 08/25/2013] [Indexed: 12/19/2022]
Abstract
Animal models for studying human disease are essential to the continuing evolution of medicine. Rodent models are attractive for the obvious similarities in development and genetic makeup compared with humans, but have cost and technical limitations. The zebrafish (Danio rerio) represents an ideal alternative vertebrate model of human disease because of its high conservation of genetic information and physiological processes, inexpensive maintenance, and optical clarity facilitating direct observation. This review highlights recent advances in understanding genetic disease states associated with the dynamic organelle, the mitochondrion, using the zebrafish. Mitochondrial diseases that have been replicated in the zebrafish include those affecting the nervous and cardiovascular systems, as well as red blood cell function. Gene silencing techniques, including morpholino knockdown and transcription activator-like (TAL)-effector endonucleases, have been exploited to demonstrate how loss of function can induce human disease-like states in zebrafish. Moreover, modeling mitochondrial diseases has been facilitated greatly by the creation of transgenic fish with fluorescently labeled mitochondria for in vivo visualization of these structures. In addition, behavioral assays have been developed to examine changes in motor activity and sensory responses, particularly in larval stages. Zebrafish are poised to advance our understanding of the pathogenesis of human mitochondrial diseases beyond the current state of knowledge and provide a key tool in the development of novel therapeutic approaches to treat these conditions.
Collapse
Affiliation(s)
- Shelby L Steele
- Department of Pediatrics, Dalhousie University, IWK Health Centre, Halifax, Nova Scotia, Canada
| | - Sergey V Prykhozhij
- Department of Pediatrics, Dalhousie University, IWK Health Centre, Halifax, Nova Scotia, Canada
| | - Jason N Berman
- Department of Pediatrics, Dalhousie University, IWK Health Centre, Halifax, Nova Scotia, Canada.
| |
Collapse
|
38
|
Pickart MA, Klee EW. Zebrafish approaches enhance the translational research tackle box. Transl Res 2014; 163:65-78. [PMID: 24269745 DOI: 10.1016/j.trsl.2013.10.007] [Citation(s) in RCA: 36] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/07/2013] [Revised: 10/24/2013] [Accepted: 10/28/2013] [Indexed: 01/08/2023]
Abstract
During the past few decades, zebrafish (Danio rerio) have been a workhorse for developmental biology and genetics. Concurrently, zebrafish have proved highly accessible and effective for translational research by providing a vertebrate animal model useful for gene discovery, disease modeling, chemical genetic screening, and other medically relevant studies. Key resources such as an annotated and complete genome sequence, and diverse tools for genetic manipulation continue to spur broad use of zebrafish. Thus, the purpose of this introductory review is to provide a window into the unique characteristics and diverse uses of zebrafish and to highlight in particular the increasing relevance of zebrafish as a translational animal model. This is accomplished by reviewing broad considerations of anatomic and physiological conservation, approaches for disease modeling and creation, general laboratory methods, genetic tools, genome conservation, and diverse opportunities for functional validation. Additional commentary throughout the review also guides the reader to the 4 new reviews found elsewhere in this special issue that showcase the many unique ways the zebrafish is improving understanding of renal regeneration, mitochondrial disease, dyslipidemia, and aging, for example. With many other possible approaches and a rapidly increasing number of medically relevant reports, zebrafish approaches enhance significantly the tools available for translational research and are actively improving the understanding of human disease.
Collapse
Affiliation(s)
| | - Eric W Klee
- Mayo Clinic, College of Medicine, Rochester, Minn
| |
Collapse
|
39
|
Mortalin and DJ-1 coordinately regulate hematopoietic stem cell function through the control of oxidative stress. Blood 2014; 123:41-50. [DOI: 10.1182/blood-2013-06-508333] [Citation(s) in RCA: 48] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/03/2023] Open
Abstract
Key Points
Mitochondrial heat shock protein, mortalin, is essential for the maintenance of HSCs via the control of oxidative stress. Mortalin directly interact with DJ-1 to regulate ROS levels in the mitochondria of HSCs.
Collapse
|
40
|
Cherian S, Bagg A. The genetics of the myelodysplastic syndromes: Classical cytogenetics and recent molecular insights. Hematology 2013; 11:1-13. [PMID: 16522543 DOI: 10.1080/10245330500276691] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/07/2023] Open
Abstract
Myelodysplastic syndromes (MDS) are a complex group of clonal hematopoietic disorders with an attendant diverse array of associated genetic changes. Conventional cytogenetics plays a prominent and well-established role in determining the contemporary diagnosis and prognosis of these disorders. More recently, molecular approaches have been useful in further characterizing this group of diseases, albeit in a largely experimental context, with the detection of changes at the single gene level including mutations, amplification and epigenetic phenomena. Nevertheless, we remain largely ignorant of the genetic underpinnings of MDS. Here we briefly review the established role of cytogenetics in MDS, and emphasize recent advances in unraveling the genetics of MDS, with a view towards how such findings might facilitate our ability to understand, diagnose and treat these disorders in a more rational manner.
Collapse
Affiliation(s)
- Sindhu Cherian
- University of Washington Medical Center, Department of Laboratory Medicine, Seattle, 98195, USA.
| | | |
Collapse
|
41
|
Komrokji RS, Padron E, Ebert BL, List AF. Deletion 5q MDS: molecular and therapeutic implications. Best Pract Res Clin Haematol 2013; 26:365-75. [PMID: 24507813 DOI: 10.1016/j.beha.2013.10.013] [Citation(s) in RCA: 40] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Abstract
Heterozygous, interstitial deletions of chromosome 5q are the most common cytogenetic abnormality in myelodysplastic syndromes (MDS). This chromosomal abnormality is associated with a consistent clinical phenotype, the 5q- syndrome, in a subset of patients, and therapeutic sensitivity to the drug lenalidomide. No genes on chromosome 5q undergo recurrent homozygous inactivation in MDS patients. Instead, haploinsufficiency for key genes powerfully alters hematopoiesis, leading to the MDS phenotype in patients with del(5q). Haploinsufficiency for the RPS14 gene leads to activation of the p53 pathway and the macrocytic anemia characteristic of this disorder, and loss of p53 rescues erythropoiesis and facilitates clonal progression. Other genes, as well as miR-145 and miR-146a, contribute to aberrant megakaryopoiesis and a selective advantage for the del(5q) clone. The integrated effects of haploinsufficiency for these key genes, in aggregate, lead to the full phenotype of the disorder.
Collapse
Affiliation(s)
- Rami S Komrokji
- H Lee Moffitt Cancer Center and Research Institute, 12902 Magnolia Drive, Tampa, FL 33612, USA.
| | - Eric Padron
- H Lee Moffitt Cancer Center and Research Institute, 12902 Magnolia Drive, Tampa, FL 33612, USA.
| | - Benjamin L Ebert
- Brigham and Women's Hospital, Karp 5.210, 1 Blackfan Circle, Boston, MA 02115, USA.
| | - Alan F List
- H Lee Moffitt Cancer Center and Research Institute, 12902 Magnolia Drive, Tampa, FL 33612, USA.
| |
Collapse
|
42
|
Invernizzi R, Travaglino E, Della Porta MG, Gallì A, Malcovati L, Rosti V, Bergamaschi G, Erba BG, Bellistri F, Bastia R, Santambrogio P, Levi S, Cazzola M. Effects of mitochondrial ferritin overexpression in normal and sideroblastic erythroid progenitors. Br J Haematol 2013; 161:726-737. [PMID: 23573868 DOI: 10.1111/bjh.12316] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2012] [Accepted: 02/04/2013] [Indexed: 01/06/2023]
Abstract
In myelodysplastic syndromes with ring sideroblasts (MDS-RS), the iron deposited in the mitochondria of RS is present in the form of mitochondrial ferritin (FTMT), but it is unknown whether FTMT overexpression is the cause or the result of mitochondrial iron deposition. Lentivirus FTMT-transduced CD34(+) bone marrow cells from seven healthy donors and CD34(+) cells from 24 patients with MDS-RS were cultured according to a procedure that allowed the expansion of high numbers of erythroid progenitors. These cells were used to investigate the possible influence of experimentally-induced FTMT overexpression on normal erythropoiesis and the functional effects of FTMT in sideroblastic erythropoiesis. In MDS-RS progenitors, FTMT overexpression was associated with reduced cytosolic ferritin levels, increased surface transferrin receptor expression and reduced cell proliferation; FTMT effects were independent of SF3B1 mutation status. Similarly, FTMT overexpressing normal erythroid progenitors were characterized by reduced cytosolic ferritin content and increased CD71 expression, and also by higher apoptotic rate in comparison with the FTMT- controls. Significantly lower levels of STAT5 phosphorylation following erythropoietin stimulation were found in both sideroblastic and normal FTMT(+) erythroid cells compared to the FTMT- cells. In conclusion, experimental overexpression of FTMT may modify mitochondrial iron availability and lead to ineffective erythropoiesis.
Collapse
Affiliation(s)
- Rosangela Invernizzi
- Department of Internal Medicine, University of Pavia Medical School and Fondazione IRCCS Policlinico San Matteo, Pavia, Italy
| | - Erica Travaglino
- Department of Haematology Oncology, University of Pavia Medical School and Fondazione IRCCS Policlinico San Matteo, Pavia, Italy
| | - Matteo G Della Porta
- Department of Haematology Oncology, University of Pavia Medical School and Fondazione IRCCS Policlinico San Matteo, Pavia, Italy
| | - Anna Gallì
- Department of Haematology Oncology, University of Pavia Medical School and Fondazione IRCCS Policlinico San Matteo, Pavia, Italy
| | - Luca Malcovati
- Department of Haematology Oncology, University of Pavia Medical School and Fondazione IRCCS Policlinico San Matteo, Pavia, Italy
| | - Vittorio Rosti
- Unit of Clinical Epidemiology, Fondazione IRCCS Policlinico San Matteo, Pavia, Italy
| | - Gaetano Bergamaschi
- Department of Internal Medicine, University of Pavia Medical School and Fondazione IRCCS Policlinico San Matteo, Pavia, Italy
| | - Benedetta G Erba
- Proteomics of Iron Metabolism Unit, Division of Neuroscience, San Raffaele Scientific Institute, Milano, Italy
| | - Francesca Bellistri
- Department of Internal Medicine, University of Pavia Medical School and Fondazione IRCCS Policlinico San Matteo, Pavia, Italy
| | - Raffaella Bastia
- Department of Internal Medicine, University of Pavia Medical School and Fondazione IRCCS Policlinico San Matteo, Pavia, Italy
| | - Paolo Santambrogio
- Proteomics of Iron Metabolism Unit, Division of Neuroscience, San Raffaele Scientific Institute, Milano, Italy
| | - Sonia Levi
- Proteomics of Iron Metabolism Unit, Division of Neuroscience, San Raffaele Scientific Institute, Milano, Italy
- Vita-Salute San Raffaele University, Milano, Italy
| | - Mario Cazzola
- Department of Haematology Oncology, University of Pavia Medical School and Fondazione IRCCS Policlinico San Matteo, Pavia, Italy
| |
Collapse
|
43
|
Hosen MJ, Vanakker OM, Willaert A, Huysseune A, Coucke P, De Paepe A. Zebrafish models for ectopic mineralization disorders: practical issues from morpholino design to post-injection observations. Front Genet 2013; 4:74. [PMID: 23760765 PMCID: PMC3669896 DOI: 10.3389/fgene.2013.00074] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2012] [Accepted: 04/15/2013] [Indexed: 01/06/2023] Open
Abstract
Zebrafish (ZF, Danio rerio) has emerged as an important and popular model species to study different human diseases. Key regulators of skeletal development and calcium metabolism are highly conserved between mammals and ZF. The corresponding orthologs share significant sequence similarities and an overlap in expression patterns when compared to mammals, making ZF a potential model for the study of mineralization-related disorders and soft tissue mineralization. To characterize the function of early mineralization-related genes in ZF, these genes can be knocked down by injecting morpholinos into early stage embryos. Validation of the morpholino needs to be performed and the concern of aspecific effects can be addressed by applying one or more independent techniques to knock down the gene of interest. Post-injection assessment of early mineralization defects can be done using general light microscopy, calcein staining, Alizarin red staining, Alizarin red-Alcian blue double staining, and by the use of transgenic lines. Examination of general molecular defects can be done by performing protein and gene expression analysis, and more specific processes can be explored by investigating ectopic mineralization-related mechanisms such as apoptosis and mitochondrial dysfunction. In this paper, we will discuss all details about the aforementioned techniques; shared knowledge will be very useful for the future investigation of ZF models for ectopic mineralization disorders and to understand the underlying pathways involved in soft tissue calcification.
Collapse
Affiliation(s)
- Mohammad Jakir Hosen
- Center for Medical Genetics, Ghent University Hospital Ghent, Belgium ; Department of Genetic Engineering and Biotechnology, Shahjalal University of Science and Technology Sylhet, Bangladesh
| | | | | | | | | | | |
Collapse
|
44
|
Pérez-Sánchez J, Borrel M, Bermejo-Nogales A, Benedito-Palos L, Saera-Vila A, Calduch-Giner JA, Kaushik S. Dietary oils mediate cortisol kinetics and the hepatic mRNA expression profile of stress-responsive genes in gilthead sea bream (Sparus aurata) exposed to crowding stress. Implications on energy homeostasis and stress susceptibility. COMPARATIVE BIOCHEMISTRY AND PHYSIOLOGY D-GENOMICS & PROTEOMICS 2013; 8:123-30. [PMID: 23466468 DOI: 10.1016/j.cbd.2013.02.001] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/21/2012] [Revised: 02/05/2013] [Accepted: 02/05/2013] [Indexed: 10/27/2022]
Abstract
Juveniles of gilthead sea bream were fed with plant protein-based diets with fish oil (FO diet) or vegetable oils (66VO diet) as dietary lipid sources. No differences in growth performance were found between both groups, and fish with an average body mass of 65-70 g were crowded (90-100 kg/m(3)) to assess the stress response within the 72 h after the onset of stressor. The rise in plasma cortisol and glucose levels was higher in stressed fish of group 66VO (66VO-S) than in FO group (FO-S), but the former stressed group regained more quickly the cortisol resting values of the corresponding non-stressed diet group. The cell-tissue repair response represented by derlin-1, 75 kDa glucose-regulated protein and 170 kDa glucose-regulated protein was triggered at a lower level in 66VO-S than in FO-S fish. This occurred in concert with a long-lasting up-regulation of glucocorticoid receptors, antioxidant enzymes, enzyme subunits of the mitochondrial respiratory chain, and enzymes involved in tissue fatty acid uptake and β-oxidation. This gene expression pattern allows a metabolic phenotype that is prone to "high power" mitochondria, which would support the replacement of fish oil with vegetable oils when theoretical requirements in essential fatty acids for normal growth are met by diet.
Collapse
Affiliation(s)
- Jaume Pérez-Sánchez
- Nutrigenomics and Fish Growth Endocrinology, Instituto de Acuicultura Torre de la Sal, IATS-CSIC, Castellón, Spain.
| | | | | | | | | | | | | |
Collapse
|
45
|
Xu J, Du L, Wen Z. Myelopoiesis during Zebrafish Early Development. J Genet Genomics 2012; 39:435-42. [DOI: 10.1016/j.jgg.2012.06.005] [Citation(s) in RCA: 42] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2012] [Revised: 06/21/2012] [Accepted: 06/21/2012] [Indexed: 11/28/2022]
|
46
|
Vu MT, Zhai P, Lee J, Guerra C, Liu S, Gustin MC, Silberg JJ. The DNLZ/HEP zinc-binding subdomain is critical for regulation of the mitochondrial chaperone HSPA9. Protein Sci 2012; 21:258-67. [PMID: 22162012 DOI: 10.1002/pro.2012] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2011] [Revised: 11/04/2011] [Accepted: 11/27/2011] [Indexed: 01/20/2023]
Abstract
Human mitochondrial DNLZ/HEP regulates the catalytic activity and solubility of the mitochondrial hsp70 chaperone HSPA9. Here, we investigate the role that the DNLZ zinc-binding and C-terminal subdomains play in regulating HSPA9. We show that truncations lacking portions of the zinc-binding subdomain (ZBS) do not affect the solubility of HSPA9 or its ATPase domain, whereas those containing the ZBS and at least 10 residues following this subdomain enhance chaperone solubility. Binding measurements further show that DNLZ requires its ZBS to form a stable complex with the HSPA9 ATPase domain, and ATP hydrolysis measurements reveal that the ZBS is critical for full stimulation of HSPA9 catalytic activity. We also examined if DNLZ is active in vivo. We found that DNLZ partially complements the growth of Δzim17 Saccharomyces cerevisiae, and we discovered that a Zim17 truncation lacking a majority of the C-terminal subdomain strongly complements growth like full-length Zim17. These findings provide direct evidence that human DNLZ is a functional ortholog of Zim17. In addition, they implicate the pair of antiparallel β-strands that coordinate zinc in Zim17/DNLZ-type proteins as critical for binding and regulating hsp70 chaperones.
Collapse
Affiliation(s)
- Michael T Vu
- Department of Biochemistry and Cell Biology, Rice University, Houston, TX 77251, USA
| | | | | | | | | | | | | |
Collapse
|
47
|
Fan GC. Role of heat shock proteins in stem cell behavior. PROGRESS IN MOLECULAR BIOLOGY AND TRANSLATIONAL SCIENCE 2012; 111:305-22. [PMID: 22917237 DOI: 10.1016/b978-0-12-398459-3.00014-9] [Citation(s) in RCA: 45] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/01/2023]
Abstract
Stress response is well appreciated to induce the expression of heat shock proteins (Hsps) in the cell. Numerous studies have demonstrated that Hsps function as molecular chaperones in the stabilization of intracellular proteins, repairing damaged proteins, and assisting in protein translocation. Various kinds of stem cells (embryonic stem cells, adult stem cells, or induced pluripotent stem cells) have to maintain their stemness and, under certain circumstances, undergo stress. Therefore, Hsps should have an important influence on stem cells. Actually, numerous studies have indicated that some Hsps physically interact with a number of transcription factors as well as intrinsic and extrinsic signaling pathways. Importantly, alterations in Hsp expression have been demonstrated to affect stem cell behavior including self-renewal, differentiation, sensitivity to environmental stress, and aging. This chapter summarizes recent findings related to (1) the roles of Hsps in maintenance of stem cell dormancy, proliferation, and differentiation; (2) the expression signature of Hsps in embryonic/adult stem cells and differentiated stem cells; (3) the protective roles of Hsps in transplanted stem cells; and (4) the possible roles of Hsps in stem cell aging.
Collapse
Affiliation(s)
- Guo-Chang Fan
- Department of Pharmacology and Cell Biophysics, University of Cincinnati College of Medicine, Cincinnati, Ohio, USA
| |
Collapse
|
48
|
Hopfer O, Nolte F, Mossner M, Komor M, Kmetsch A, Benslasfer O, Reißmann M, Nowak D, Hoelzer D, Thiel E, Hofmann WK. Epigenetic dysregulation of GATA1 is involved in myelodysplastic syndromes dyserythropoiesis. Eur J Haematol 2011; 88:144-53. [DOI: 10.1111/j.1600-0609.2011.01715.x] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
|
49
|
|
50
|
Keightley MC, Layton JE, Hayman JW, Heath JK, Lieschke GJ. Mediator subunit 12 is required for neutrophil development in zebrafish. PLoS One 2011; 6:e23845. [PMID: 21901140 PMCID: PMC3162013 DOI: 10.1371/journal.pone.0023845] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2011] [Accepted: 07/25/2011] [Indexed: 11/19/2022] Open
Abstract
Hematopoiesis requires the spatiotemporal organization of regulatory factors to successfully orchestrate diverse lineage specificity from stem and progenitor cells. Med12 is a regulatory component of the large Mediator complex that enables contact between the general RNA polymerase II transcriptional machinery and enhancer bound regulatory factors. We have identified a new zebrafish med12 allele, syr, with a single missense mutation causing a valine to aspartic acid change at position 1046. Syr shows defects in hematopoiesis, which predominantly affect the myeloid lineage. Syr has identified a hematopoietic cell-specific requirement for Med12, suggesting a new role for this transcriptional regulator.
Collapse
Affiliation(s)
- Maria-Cristina Keightley
- Australian Regenerative Medicine Institute, Monash University, Clayton, Victoria, Australia
- Cancer and Haematology Division, Walter and Eliza Hall Institute of Medical Research, Parkville, Victoria, Australia
| | - Judith E. Layton
- Cancer and Haematology Division, Walter and Eliza Hall Institute of Medical Research, Parkville, Victoria, Australia
| | - John W. Hayman
- Australian Regenerative Medicine Institute, Monash University, Clayton, Victoria, Australia
- Cancer and Haematology Division, Walter and Eliza Hall Institute of Medical Research, Parkville, Victoria, Australia
| | - Joan K. Heath
- Colon Molecular and Cell Biology Laboratory, Melbourne Branch, Ludwig Institute for Cancer Research, The Royal Melbourne Hospital, Parkville, Victoria, Australia
| | - Graham J. Lieschke
- Australian Regenerative Medicine Institute, Monash University, Clayton, Victoria, Australia
- Cancer and Haematology Division, Walter and Eliza Hall Institute of Medical Research, Parkville, Victoria, Australia
- Department of Medical Biology, University of Melbourne, Parkville, Victoria, Australia
| |
Collapse
|