Musashi-2

Last updated
MSI2
Musashi2 protein in homolog 2 in Homo sapiens.png
Identifiers
Aliases MSI2 , MSI2H, musashi RNA binding protein 2, Musashi2, Musashi-2
External IDs OMIM: 607897 MGI: 1923876 HomoloGene: 62199 GeneCards: MSI2
Orthologs
SpeciesHumanMouse
Entrez
Ensembl
UniProt
RefSeq (mRNA)

NM_138962
NM_170721
NM_001322250
NM_001322251

NM_001201341
NM_054043
NM_001363194
NM_001363195
NM_001373923

Contents

RefSeq (protein)

NP_001309179
NP_001309180
NP_620412
NP_733839

NP_001188270
NP_473384
NP_001350123
NP_001350124
NP_001360852

Location (UCSC) Chr 17: 57.26 – 57.68 Mb Chr 11: 88.23 – 88.61 Mb
PubMed search [3] [4]
Wikidata
View/Edit Human View/Edit Mouse
Musashi-2 protein in homolog 2 in Homo sapiens. Musashi2 protein in homolog 2 in Homo sapiens.png
Musashi-2 protein in homolog 2 in Homo sapiens.

Musashi-2, also known as Musashi RNA binding protein 2, is a protein that in humans is encoded by the MSI2 gene. [5] Like its homologue musashi-1 (MSI1), it is an RNA-binding protein involved in stemness.

Expression

There are two homologue genes found in mammals, called musashi1 (MSI1) and musashi-2 (MSI2). Musashi-2 is an RNA-binding protein expressed in neuronal progenitor cells, including stem cells, and both normal and leukemic blood cells. [6] [7]

Musashi-2 also appears to be expressed in stem cells and in a wide variety of tissues, including the bulge region of the hair follicle, immature pancreatic β-cells and neural progenitor cells. [6] Amongst the last ones, MSI2 is expressed in early stages of development, in the ventricular and subventricular zone, [8] in cells of the astrocyte lineage. It was there that it was first discovered. [6] Within the hematopoietic system, MSI2 is highly expressed in the most primitive progenitors, [6] [9] in stem cell compartments, [7] and its overexpression has been found in myeloid leukemia cell lines. [7] In neural cell lines, MSI2 protein, as well as its homologue MSI1, is exclusively located in the cytoplasm. [8]

In humans, the MSI2 gene is located in chromosome 17q23.2. [10] and has a sequence length of 1,414bp of which 987bp are encoded. [11] In mice, MSI2 has been found to be in 11qB5-C [8] and BC169841 in the African clawed frog (Xenopus laevis). [9] There are two different isoforms of MSI2 expressed by embryonic stem cells that result from alternative splicing, isoform 1 and isoform 2. The first one is the larger canonical isoform, and the second one is the shorter, splice-variant Isoform.

Function

This gene encodes an RNA-binding protein that is a member of the Musashi protein family. The encoded protein is translational regulator that targets genes involved in development and cell cycle regulation. Mutations in this gene are associated with poor prognosis in certain types of cancers. This gene has also been shown to be rearranged in certain cancer cells. The first musashi (abbreviation MSI) gene was first discovered in Drosophila and then later identified in other eukaryotic species.

MSI2 is involved in organismal development. [12] As with the rest of Musashi family RNA-binding proteins, MSI2 is linked to tissue stem cells and has an influence in asymmetric cell division, germ and somatic stem cell function and cell fate determination in a variety of tissues. [7]

As an RNA-binding protein, MSI2 is acts as a translational inhibitor. [9] Through this molecular mechanism, MSI2 contributes in more than one vital aspect, as in the development of the nervous system, regulation of the Hematopoietic stem cell (HSC) compartment, or the self-renewal and pluripotency of embryonic stem cells. MSI2 takes part in a high number of pathways related to the self-renewal of some stem cells. However, it is not only focused in one specific type. Depending on the tissue where it is located, it develops different functions.

Embryonic stem cells

MSI2 belongs to the RNA-processing group of proteins which are associated with the transcription factor SOX2 during the early stages of differentiation. SOX2 is known to be essential during embryogenesis and in the self-renewal and pluripotency of embryonic stem cells. MSI2 has a high influence on it too, since the gain or loss of self-renewal capacity and the extent of differentiation depends on MSI2 levels. Although both of the isoforms of this protein are needed to the maintenance of the self-renewal, they are different on a functional way and they play different roles in some aspects of the process. For example, only isoform 1 expression is related to the cloning efficiency of embryonic stem cells. [12]

Neural progenitor stem cells

In a similar way to MSI1, MSI2 is also active in the proliferation of pluripotent neural precursors cells of the embryo, during which both MSI1 and MSI2 are strongly co-expressed. Moreover, MSI1 and MSI2 regulate the multiplication and maintenance of a specific group inside of neural precursors cells: CNS (central neural system) stem cells populations. Therefore, MSI2 plays a significant role in the development and maintenance of CNS stem cells through post-transcriptional gene regulation. [7]

Hematopoiesis

MSI2 is present in blood cells, in which its expression is situated in the hematopoietic system, more commonly in the most primitive cells. These are the LSK cells, which are composed by long-term hematopoietic stem cells (LT-HSCs), short-term HSCs (ST-HCSs) and multipotent progenitors (MPPs). [6]

Self-renewal and differentiation processes in hematopoietic stem cells need to be highly regulated in order to maintain homeostasis and to avoid the growing of blood cell malignancies. It is this point is where Musashi-2 interferes. [9] Therefore, MSI2’s function in HSCs consists of regulating their proliferation and differentiation. Therefore, a decreasing on the level of MSI2 induces a reduction in the number of more primitive progenitors of HSCs. [6]

Clinical significance

As Musashi-2 is involved in the generation of hematopoietic cells, it is also linked with cancer pathologies:

Myeloid leukemia

It has been found that MSI2 plays an important role in myeloid leukemia. In both of chronic myelogenous leukemia (CML) and acute myeloid leukemia (AML), MSI2 regulates hematopoietic stem cell proliferation and does not allow the differentiation of its gene expression. [7]

Chronic myelogenous leukemia

Chronic myelogenous leukemia (CML) progresses from the initial phase, where differentiated myeloid cells are accumulated, to the accelerated phase, where the expansion of these cells increases, and it ends with the blast crisis phase. It has been found that MSI2 participates together with BCR-ABL gene to stimulate the progress to the aggressive phase. [7] The first evidence to consider its role in this phase is its high concentration compared with the first phase of the disease. One of the functions of the MSI2 is to regulate the expression of NUMB, causing its inhibition. [13] Therefore, the function of the MSI2 in this disease is being studied together with Numb expression. However, while Numb is overexpressed during the chronic phase and decreases in the blast one, Musashi starts to be overexpressed in the last fatal phase of CML. [14] The high expression of MSI2 interrupts the cellular differentiation and allows the expansion of immature leukemic cells causing the progress to the deadly phase. [14]

Acute myeloid leukemia

As acute myeloid leukemia (AML) has a similar behavior to the aggressive phase of the CML, MSI2’s role is similar there as well. It has been found that MSI2 is overexpressed in this type of leukemia and its activity is related with Numb consequently. Moreover, the high expression of MSI2 is related with a poor clinical outcome. [6] In order to prove this, it has been demonstrated that with MSI’s knockdown leads to a rising apoptosis and differentiation and to a decreasing proliferation. [14] As a result, patients that develop leukemia without a high expression of MSI2 have a better prognostic.

Diagnostic and therapeutic applications

MIS2 is a potential cancer biomarker as well as a drug target. [15]

See also

Related Research Articles

<span class="mw-page-title-main">Haematopoiesis</span> Formation of blood cellular components

Haematopoiesis is the formation of blood cellular components. All cellular blood components are derived from haematopoietic stem cells. In a healthy adult person, approximately 1011–1012 new blood cells are produced daily in order to maintain steady state levels in the peripheral circulation.

<span class="mw-page-title-main">Hematopoietic stem cell</span> Stem cells that give rise to other blood cells

Hematopoietic stem cells (HSCs) are the stem cells that give rise to other blood cells. This process is called haematopoiesis. In vertebrates, the very first definitive HSCs arise from the ventral endothelial wall of the embryonic aorta within the (midgestational) aorta-gonad-mesonephros region, through a process known as endothelial-to-hematopoietic transition. In adults, haematopoiesis occurs in the red bone marrow, in the core of most bones. The red bone marrow is derived from the layer of the embryo called the mesoderm.

<span class="mw-page-title-main">CD135</span> Protein-coding gene in the species Homo sapiens

Cluster of differentiation antigen 135 (CD135) also known as fms like tyrosine kinase 3 (FLT-3), receptor-type tyrosine-protein kinase FLT3, or fetal liver kinase-2 (Flk2) is a protein that in humans is encoded by the FLT3 gene. FLT3 is a cytokine receptor which belongs to the receptor tyrosine kinase class III. CD135 is the receptor for the cytokine Flt3 ligand (FLT3L).

<span class="mw-page-title-main">Stem cell factor</span> Mammalian protein found in Homo sapiens

Stem cell factor is a cytokine that binds to the c-KIT receptor (CD117). SCF can exist both as a transmembrane protein and a soluble protein. This cytokine plays an important role in hematopoiesis, spermatogenesis, and melanogenesis.

<span class="mw-page-title-main">RUNX1</span> Protein-coding gene in the species Homo sapiens

Runt-related transcription factor 1 (RUNX1) also known as acute myeloid leukemia 1 protein (AML1) or core-binding factor subunit alpha-2 (CBFA2) is a protein that in humans is encoded by the RUNX1 gene.

<span class="mw-page-title-main">HOXA9</span> Protein-coding gene in humans

Homeobox protein Hox-A9 is a protein that in humans is encoded by the HOXA9 gene.

<span class="mw-page-title-main">NUMB (gene)</span>

Protein numb homolog is a protein that in humans is encoded by the NUMB gene. The protein encoded by this gene plays a role in the determination of cell fates during development. The encoded protein, whose degradation is induced in a proteasome-dependent manner by MDM2, is a membrane-bound protein that has been shown to associate with EPS15, LNX1, and NOTCH1. Four transcript variants encoding different isoforms have been found for this gene.

<span class="mw-page-title-main">CEBPA</span> Protein-coding gene in the species Homo sapiens

CCAAT/enhancer-binding protein alpha is a protein encoded by the CEBPA gene in humans. CCAAT/enhancer-binding protein alpha is a transcription factor involved in the differentiation of certain blood cells. For details on the CCAAT structural motif in gene enhancers and on CCAAT/Enhancer Binding Proteins see the specific page.

<span class="mw-page-title-main">Homeobox A10</span> Protein-coding gene in humans

Homeobox protein Hox-A10 is a protein that in humans is encoded by the HOXA10 gene.

<i>ERG</i> (gene) Protein-coding gene in the species Homo sapiens

ERG is an oncogene. ERG is a member of the ETS family of transcription factors. The ERG gene encodes for a protein, also called ERG, that functions as a transcriptional regulator. Genes in the ETS family regulate embryonic development, cell proliferation, differentiation, angiogenesis, inflammation, and apoptosis.

<span class="mw-page-title-main">HOXB6</span> Protein-coding gene in the species Homo sapiens

Homeobox protein Hox-B6 is a protein that in humans is encoded by the HOXB6 gene.

<span class="mw-page-title-main">MECOM</span>

MDS1 and EVI1 complex locus protein EVI1 (MECOM) also known as ecotropic virus integration site 1 protein homolog (EVI-1) or positive regulatory domain zinc finger protein 3 (PRDM3) is a protein that in humans is encoded by the MECOM gene. EVI1 was first identified as a common retroviral integration site in AKXD murine myeloid tumors. It has since been identified in a plethora of other organisms, and seems to play a relatively conserved developmental role in embryogenesis. EVI1 is a nuclear transcription factor involved in many signaling pathways for both coexpression and coactivation of cell cycle genes.

<span class="mw-page-title-main">ID4</span> Protein-coding gene in humans

ID4 is a protein coding gene. In humans, it encodes for the protein known as DNA-binding protein inhibitor ID-4. This protein is known to be involved in the regulation of many cellular processes during both prenatal development and tumorigenesis. This is inclusive of embryonic cellular growth, senescence, cellular differentiation, apoptosis, and as an oncogene in angiogenesis.

<span class="mw-page-title-main">Musashi-1</span>

RNA-binding protein Musashi homolog 1 also known as Musashi-1 is a protein that in humans is encoded by the MSI1 gene.

<span class="mw-page-title-main">PHF6</span> Protein-coding gene in the species Homo sapiens

PHD finger protein 6 is a protein that in humans is encoded by the PHF6 gene.

<span class="mw-page-title-main">HOXA6</span>

Homeobox protein Hox-A6 is a protein that in humans is encoded by the HOXA6 gene.

BAALC is a gene that codes for the: brain and acute leukemia cytoplasmic protein. The official symbol (BAALC) and official name is maintained by the HGNC. The function of BAALC is not fully understood yet but has been suggested to have synaptic roles involving the post synaptic lipid raft. Lipid rafts are microdomains that are enriched with cholesterol and sphingolipids, lipid raft functions include membrane trafficking, signal processing, and regulation of the actin cytoskeleton. The postsynaptic lipid raft possesses many proteins and is one of the major sites for signal processing and postsynaptic density (PSD). Along with its involvement in the post synaptic lipid rafts, BAALC expression has been associated with Acute Lymphoblastic Leukemia and Acute Myeloid Leukemia.

Stem cell markers are genes and their protein products used by scientists to isolate and identify stem cells. Stem cells can also be identified by functional assays. Below is a list of genes/protein products that can be used to identify various types of stem cells, or functional assays that do the same. The initial version of the list below was obtained by mining the PubMed database as described in

mir-223

In molecular biology MicroRNA-223 (miR-223) is a short RNA molecule. MicroRNAs function to regulate the expression levels of other genes by several mechanisms. miR-223 is a hematopoietic specific microRNA with crucial functions in myeloid lineage development. It plays an essential role in promoting granulocytic differentiation while also being associated with the suppression of erythrocytic differentiation. miR-223 is commonly repressed in hepatocellular carcinoma and leukemia. Higher expression levels of miRNA-223 are associated with extranodal marginal-zone lymphoma of mucosa-associated lymphoid tissue of the stomach and recurrent ovarian cancer. In some cancers the microRNA-223 down-regulation is correlated with higher tumor burden, disease aggressiveness, and poor prognostic factors. MicroRNA-223 is also associated with rheumatoid arthritis, sepsis, type 2 diabetes, and hepatic ischemia.

AI-10-49 is a small molecule inhibitor of leukemic oncoprotein CBFβ-SMHHC developed by the laboratory of John Bushweller with efficacy demonstrated by the laboratories of Lucio H. Castilla and Monica Guzman. AI-10-49 allosterically binds to CBFβ-SMMHC and disrupts protein-protein interaction between CBFβ-SMMHC and tumor suppressor RUNX1. This inhibitor is under development as an anti-leukemic drug.

References

  1. 1 2 3 GRCh38: Ensembl release 89: ENSG00000153944 - Ensembl, May 2017
  2. 1 2 3 GRCm38: Ensembl release 89: ENSMUSG00000069769 - Ensembl, May 2017
  3. "Human PubMed Reference:". National Center for Biotechnology Information, U.S. National Library of Medicine.
  4. "Mouse PubMed Reference:". National Center for Biotechnology Information, U.S. National Library of Medicine.
  5. "Entrez Gene: Musashi RNA binding protein 2".
  6. 1 2 3 4 5 6 7 de Andrés-Aguayo L, Varas F, Graf T (July 2012). "Musashi 2 in hematopoiesis". Current Opinion in Hematology. 19 (4): 268–72. doi:10.1097/MOH.0b013e328353c778. PMID   22517588. S2CID   205827403.
  7. 1 2 3 4 5 6 7 Kharas MG, Lengner CJ, Al-Shahrour F, Bullinger L, Ball B, Zaidi S, Morgan K, Tam W, Paktinat M, Okabe R, Gozo M, Einhorn W, Lane SW, Scholl C, Fröhling S, Fleming M, Ebert BL, Gilliland DG, Jaenisch R, Daley GQ (August 2010). "Musashi-2 regulates normal hematopoiesis and promotes aggressive myeloid leukemia" (PDF). Nature Medicine. 16 (8): 903–8. doi:10.1038/nm.2187. hdl:1721.1/73937. PMC   3090658 . PMID   20616797.
  8. 1 2 3 Sakakibara S, Nakamura Y, Satoh H, Okano H (October 2001). "Rna-binding protein Musashi2: developmentally regulated expression in neural precursor cells and subpopulations of neurons in mammalian CNS". The Journal of Neuroscience. 21 (20): 8091–107. doi:10.1523/JNEUROSCI.21-20-08091.2001. PMC   6763847 . PMID   11588182.
  9. 1 2 3 4 de Andrés-Aguayo L, Varas F, Kallin EM, Infante JF, Wurst W, Floss T, Graf T (July 2011). "Musashi 2 is a regulator of the HSC compartment identified by a retroviral insertion screen and knockout mice". Blood. 118 (3): 554–64. doi: 10.1182/blood-2010-12-322081 . PMID   21613258.
  10. "Gene Symbol Report | HUGO Gene Nomenclature Committee". www.genenames.org. Archived from the original on 2013-09-17.
  11. "ENA Browser".
  12. 1 2 Wuebben EL, Mallanna SK, Cox JL, Rizzino A (2012). "Musashi2 is required for the self-renewal and pluripotency of embryonic stem cells". PLOS ONE. 7 (4): e34827. Bibcode:2012PLoSO...734827W. doi: 10.1371/journal.pone.0034827 . PMC   3319613 . PMID   22496868.
  13. Ito T, Kwon HY, Zimdahl B, Congdon KL, Blum J, Lento WE, Zhao C, Lagoo A, Gerrard G, Foroni L, Goldman J, Goh H, Kim SH, Kim DW, Chuah C, Oehler VG, Radich JP, Jordan CT, Reya T (August 2010). "Regulation of myeloid leukaemia by the cell-fate determinant Musashi". Nature. 466 (7307): 765–8. Bibcode:2010Natur.466..765I. doi:10.1038/nature09171. PMC   2918284 . PMID   20639863.
  14. 1 2 3 Griner, LN; Reuther, GW (2010). "Aggressive myeloid leukemia formation is directed by the Musashi 2/Numb pathway" (PDF). Cancer Biology & Therapy. 10 (10): 979–982. doi: 10.4161/cbt.10.10.14010 . PMID   21084860. S2CID   8975004.
  15. Melo JV, Barnes DJ (June 2007). "Chronic myeloid leukaemia as a model of disease evolution in human cancer". Nature Reviews. Cancer. 7 (6): 441–53. doi:10.1038/nrc2147. PMID   17522713. S2CID   30599281.

Further reading

This article incorporates text from the United States National Library of Medicine, which is in the public domain.