User:hadrienAnne/mir-196

From Wikipedia, the free encyclopedia
mir-196 microRNA precursor family
Identifiers
Symbolmir-196
RfamRF00256
miRBaseMI0000238
miRBase familyMIPF0000031
Other data
RNA typeGene; miRNA
Domain(s)Eukaryota
GOGO:0035195 GO:0035068
SOSO:0001244
PDB structuresPDBe

miR-196 is a non-coding RNA called a microRNA that has been shown to be expressed in numerous animal species including vertebrates and invertebrates (miRBase.org) [1,2]. There are three paralogues of miR-196 in most vertebrate species : (e.g. mir-196a-1 , mir-196a-2 and mir-196b in mouse and human). Some animals such as zebrafish have five paralogues while some animals appear to have only a single copy (Ciona intestinalis) [1]. MiR-196 appears to be an olfactores specific microRNA (including urochordates and vertebrates) and has now been predicted or experimentally confirmed in a wide range of species (MIPF0000031). miR-196 appears to be expressed from intergenic regions in HOX gene clusters. The hairpin precursors are predicted based on base pairing and cross-species conservation. In this case the mature sequence is excised from the 5' arm of the hairpin.

It has been suggested that a rare SNP (rs11614913) that overlaps hsa-mir-196a2 has been found to be associated with non-small cell lung carcinoma.[2]

Species Distribution[edit]

mir-196 is highly conserved in a wide range of animals including the urochordate Ciona intestinalis and all vertebrates investigated thus far [3] (mirbase link)

Currently these are species where the gene mir-196 was sequenced : carolina anole (Anolis carolinensis), Geoffroy's spider monkey (Ateles geoffroyi), cattle (Bos taurus), common carp (Cyprinus carpio), dog (Canis familiaris), chinese hamster (Cricetulus griseus), zebrafish (Danio rerio), horse (Equus caballus), Fugu rubripes (Japanese pufferfish), chicken, Western gorilla (Gorilla gorilla), human, brown woolly monkey (Lagothrix lagotricha), gray short-tailed opossum (Monodelphis domestica), rhesus macaque (Macaca mulatta), mouse, platypus (Ornithorhynchus anatinus), Japanese killifish (Oryzias latipes), sea lamprey (Petromyzon marinus), bonobo (Pan paniscus), Bornean orangutan (Pongo pygmaeus), common chimpanzee (Pan troglodytes), rat, pig, Zebra Finch (Taeniopygia guttata), Tetraodon nigroviridis and Western clawed frog (Xenopus tropicalis).
As well as Ciona intestinalis (sea squirt), Myxine glutinosa (atlantic hagfish), Scyliorhinus canicula (catshark), lampetra planeri (river lamprey).


MiR-10 is conserved among protostomes and deuterostomes, MiR-615 is conserved among eutherian mammals, MiR-iab-4/miR-iab-8 are conserved among arthropods and are located between abd-A (Hox8) and Abd-B, MiR-4069 are located upstream of posterior Hox orthologues. In chick, the recently acquired and MiR-1732 is positioned adjacent to miR-196b in the HoxA cluster. [4]


Genomic Locations[edit]

miR-196a-1 is located on chromosome 17 at a site between HOXB9 and HOXB10 genes. [5][6], miR-196a-2 is located on chromosome 12 at a site between HOXC10 and HOXC9 genes [5][6] and miR-196b is located at a site between HOXA9 and HOXA10 genes on chromosome 7 in human and chromosome 6 in mice. [7][5] .

File:MirRna position.png
Position of mir-196 family (mir-196a-1,mir196a-2 and mir196b).


miR-196a-1 and miR-196a-2 genes transcribe the same functional mature miRNA sequence whereas miR-196b gene produces a small RNA which differs from the sequence of miR-196a by one nucleotide [6].

miRNA Gene Location mature miRNA sequence
miR-196a-1 Chromosome 17 5′-UAGGUAGUUUCAUGUUGUUGGG-3′
miR-196a-2 Chromosome 12 5′-UAGGUAGUUUCAUGUUGUUGGG-3′
miR-196b Chromosome 6/7 5′-UAGGUAGUUUCCUGUUGUUGGG-3′

Relation with other microRNA[edit]

The mir-196 family is one of eight other microRNA families located within the Hox clusters; mir-10, mir-196, mir-615, mir-993, mir-4069, mir-1991,mir-1732 and miR-iab-4/8 [4] found in invertebrates such as Drosophila melanogaster, Apis mellifera and Bombyx mori.[8] [9]
These 8 microRNA families are all located on the hox clusters and may have similar function. Hox genes encode homeodomain-containing transcription factors that are essential for embryonic development.[10]</ref>

File:The Hox clusters.png
Evolution of microRNA located of Hox clusters.

===Relation with miR-10 family=== (there is no direct relation to miR-10 aside from both being in the hox clusters... function and sequence are completely different)


Targets of miR-196[edit]

In ovo application of antagomiRs indicates a role for miR-196 in patterning the chick axial skeleton through Hox gene regulation. |journal=Proc Natl Acad Sci U S A |volume=106 |issue=44 |pages=18610–5 |year=2009 |pmid=19846767 |doi=10.1073/pnas.0910374106 |pmc=2773993}}</ref> [11]. In zebrafish, miR-196 has been shown to regulate the axial skeleton through regulation of the Retinoic Acid pathway, resulting in loss or gain of extra rib-bearing vertebrae (He, et al. Dev Biol. 2011 Sep 15;357(2):463-77. doi: 10.1016/j.ydbio.2011.07.014. Epub 2011 Jul 20.)

Down-regulation of HOXB8, HOXC8, HOXD8 and HOXA7 and also cleavage mechanism for miR-196-directed repression of HOXB has been demonstrated by cell culture experiments[12][13]. In ovo knockdown of miR-196 on chick resulted in highly significant posteriorizing transformation of the last cervical vertebra (C14)[9].

MiR-196 repressed the expression of HOXB8 by cleaving the mRNAs[14] and also regulates the expression of HOXC8, HOXD8 and HOXA7 by transcriptional inhibition [6].

HMGA2 gene is a target of miR-196a-2 which up-regulate miR-196a-2 expression and HMGA1 can down-regulate the expression of HMGA family members

Prediction targets[edit]

Several other genes have been predicted to be targeted by miR-196 [15], including S100 calcium-binding protein A9, small proline-rich protein 2C, keratin 5, CLCA family member 2 (a chloride channel regulator), cytochrome P450 (family 4, subfamily B, polypeptide 1), keratin 4, LDOC1, leukotriene A4 hydrolase, pleiotrophin, T-cell differentiation protein, tumour protein D52-like 1, visinin-like 1 and v-ETS erythroblastosis virus E26 oncogene homologue (ERG) . However, most of these target molecules of miR-196 have not been confirmed with experimental approaches

Mir-196 roles[edit]

Role in developement[edit]

miR-196 appears to play an important role in development :


  • In Drosophila, there is a conserved or possibly convergent interaction between the miR-196 homologue iab-4 and the homeotic Ubx HOX gene [16].
  • In chicken, the interaction between miR-196 and HOXB8 is implicated in the mechanism that prevents the mesoderm to do limb induction by retinoic acid [16].
  • miR-196 target at the upstream of HOXB8 and Sonic hedgehog (Shh) in the context of limb development, specifically in the hindlimb. [16].
  • The overexpressed of mir-196a induces eye anomalies in Xenopus laevis.

HMGA are nuclear architectural factors that play critical roles in a wide range of biological processes. Two variants of HMGA proteins, HMGA1 and HMGA2, have been identified[45]. Both HMGA genes are highly expressed during embryogenesis, and there is evidence that the absence of HMGA 2 protein causes growth retardation in mice .


  • HMGA1 is to regulate miR-196 expression, whereas miR-19a can control HMGA2 mRNA translation, which may affect development.

Precise levels of mir196 are required to initiate development of the pectoral appendage, to develop the correct number of pharyngeal arches, and to specify the number and identity of rostral vertebrae and ribs. We show that miR-196 can alter hox gene expression patterns and that miR-196 acts on pectoral appendage development by altering retinoic acid signaling via fine-tuning the expression of the retinoic acid receptor Rarab. [17]


In developing embryos, miR-196 acts to modulate Hoxb8 levels both within [16]. and immediately posterior to its endogenous expression domain in the paraxial mesoderm, and the detection of Hoxb8 endonucleolytic cleavage products in posterior regions. [16].


Role in morphognenesis[edit]

Although Hox genes chromosomal organization induces the graded expression of its genes through the anterior-posterior axis, this graded expression does not precisely align with the somites boundaries. The miR-196 plays a role in aligning and sharpening the expression of Hox genes through translation inhibition and mRNA cleavage. Differential miR-196 expression in hindlimb and forelimb has also been observed in chick and mouse (observed level of miR-196 20 fold greater in hindlimb than in forelimb). Because miR-196 downregulate HOXB8 which in turn induces Sonic Hedgehog, miR-196 is responsible of the differential expression of Sonic Hedgehog between the hindlimb and the forelimb. These observations ascribes the role of a safeguard for Hox genes expression to the miR-196 [16]. The role of miR-196 in axolotls tail regeneration has also been documented [18].

Role in cancer[edit]

Mir-196 can regulate cellular functions such as proliferation, apoptosis and differentiation.

Pancreatic cancer[edit]

The miRNA-196a levels are inversely correlated with survival in pancreatic adenocarcinoma patients. [19] Up-regulation of miR-196a has also been found in pancreatic cancer. Thus, miR-196a can be used to diagnose pancreatic cancer by raising the level in cells.

Breast cancer[edit]

Up-regulation of miR-196a has also been found in breast cancer. This included up-regulation of previously reported breast cancer-related miRNAs such as miR-21, miR-155, miR-191 and miR-196a[20]. The miR-196 family could suppress breast cancer cell migration and metastasis by inhibiting HOXC8 expression. [1]

Leukaemia[edit]

In leukaemia, levels of miR-10a, miR-10b and miR-196a-1 showed a correlation with HOX gene expression (for 30 patients with leukemia). As the overexpression of a specific subset of the Hox genes is known to be a hallmark for the Mixed-Lineage Leukemia, the role of mir-196 in this pathology has been investigated. The MLL gene regulates expression during stem cell differentiation [7]. In the pathological context of Mixed-Lineage Leukemia, the MLL protein is altered and induces the overexpression of mir-196. This overexpression of miR-196b was found specifically in patients with MLL-associated leukaemia (for 55 patients) [7][21] Leukemogenic MLL fusion proteins cause overexpression of miR-196b, which may be necessary for MLL fusion-mediated immortalization. miR-196b expression is significantly more up-regulated in AML than ALL and that, again, miR-196b up-regulation is negatively associated with overall survival of AML patients . Research of the expression of miR-196 in mouse's hematopoeitic progenitors has shown that miR-196 is expressed in long-term hematopoeitic stem cells (LT-HSC), short-term hematopoeitic stem cells (ST-HSC) and multipotent progenitors (MPP). Expression levels of mir-196b are the highest in ST-HSC and decrease as cells differentiates. In a physiological context, LT-HSCs become ST-HSCs, then MPP, then differentiated cells as the level of miR-196 decreases. In a physiological context, the high level of miR-196 increase survival and proliferation of ST-HSC and MPP and inhibits their differentiation [7].

  • The up regulation of mir-196b leads DNA methylation at CPg islands in the promoter regions of mir-196 .
  • High expression of miR-196b causes leukemia.

Other tumors[edit]

miR-196b is known to induce changes in myeloid development, and the co-expression of miR-196b and miR-21 reportedly block granulocyte colony-stimulating factor-induced granulopoiesis completely. [22] Highly elevated levels of miR-196a have also been detected in oesophageal adenocarcinoma. miR-196a may have growth-promoting and antiapoptotic functions. The expression of HMGA proteins is in general low or absent in normal tissues but overexpressed of HMGA proteins is observed in tumor cells of several cancers, however, it was noted that mir-196 inhibits the synthesis of protein HMGA so it could inhibit tumor progression.


Oncogenic function[edit]

miR-196 has oncogenic function in cancer, it is important to point out that miR-196 may play a tumour suppressive role as well. miR-196 has a dominant effect on the inhibition of oncogenic molecules so miR-196 will play a tumour suppressor function.


stomach cancer[edit]

The expression levels of miR-196 in the gastric mucosa are higher in healthy patients than patients with intesinal cancer. SHH is downregulation by miR-196 in intestinal metaplastic glands in tumor tissues. [23]

pancreatic cancer[edit]

miR-196a is up-regulated in pancreatic cancer. [13]

glioblastoma[edit]

miR-196a is up-regulated in glioblastoma. [13]

melanoma[edit]

miR-196a is down-regulated in melanoma, and its expression inhibits the invasion of melanoma cells by targeting HOXC8. [13]

Other roles[edit]

MiRNA-196 appears to have several important cellular functions in addition to development and malignancy. Endometriosis is a gynaecological disorder in females in which endometrial-like cells grow in areas outside the uterine cavity. [24].[25].

miR-196 may also play a role in the pathogenesis of severe congenital neutropenia (SCN), a rare haematological disorder characterized by an abnormally low number of neutrophils Gfi1 is a transcriptional repressor that is required for normal myelopoiesis. It has been shown that miR-196b is negatively regulated by Gfi1.[24]. miR-196 also has biological functions involving immunology, inflammation and virus defence. CD56+ T cells may be able to up-regulate the expression of miR-196a, showing in a study about hepatitis C virus (HCV). [26].

Overexpression of these miRNAs in infected liver cells leads to a marked attenuation of viral replication, demonstrating the importance of miRNAs in anti-viral immunity [27].

Since viruses also play a role in the pathogenesis of several tumour types, miRNAs may mediate viral–host interactions that have important implications not only for infectious diseases but also for cancer pathogenesis. The overexpression of miR-196a inhibited the proliferation of human adipose tissue-derived mesenchymal stem cells while enhancing osteogenic differentiation . [28]. Finally, miR-196a may be used to storage of blood.


Role in limb regeneration[edit]

Experiments on axolotl tail regeneration has shown that miR-196 (at least indirectly) downregulates the expression of Pax7, BMP4 and Msx1 (those genes are involved in maintaining dorsal cell identity). On the other hand, these experiments have also shown that levels of Meis2 (an atypical homeobox domain protein playing a role in defining proximo-distal identity during limb regeneration) are significantly lowered in inhibitor-196-treated axolotls. According to these experiments, Meis2 and miR-196 may play a role in axolotl limb regeneration by determining positional identity and translating the information of "how much is to be regenerated yet" [18].

HCV infection[edit]

miR-196 binding sites in the 3′-UTR of Bach1, which lead to down-regulation of Bach1 gene expression, up-regulation of HMOX1 gene expression, and down-regulation of HCV expression. .[29]

Crohn’s disease[edit]

miR-196, is overexpressed in the inflammatory intestinal epithelia of individuals with Crohn’s disease and downregulates the IRGM protective variant (c.313C) but not the risk-associated allele (c.313T). Subsequent loss of tight regulation of IRGM expression compromises control of intracellular replication of Crohn’s disease–associated adherent invasive Escherichia coli by autophagy The human IRGM is currently a poorly understood regulator of Xenophagy, a dedicated form of Autophagy that engulfs and degrades intracellular pathogens as a host-defense mechanism. [30]


miR-196 binding in the open reading frame of IRGM, an interaction whose misregulation may underly susceptibility to Crohn’s disease.[4]

References[edit]

  1. ^ a b Li Y, Zhang M, Chen H, Dong Z, Ganapathy V, Thangaraju M, Huang S (2010). "Ratio of miR-196s to HOXC8 messenger RNA correlates with breast cancer cell migration and metastasis". Cancer Res. 70 (20): 7894–904. doi:10.1158/0008-5472.CAN-10-1675. PMC 2955846. PMID 20736365.{{cite journal}}: CS1 maint: multiple names: authors list (link)
  2. ^ Hu Z, Chen J, Tian T, Zhou X, Gu H, Xu L, Zeng Y, Miao R, Jin G, Ma H, Chen Y, Shen H (2008). "Genetic variants of miRNA sequences and non-small cell lung cancer survival". J Clin Invest. 118 (7): 2600–8. doi:10.1172/JCI34934. PMC 2402113. PMID 18521189.{{cite journal}}: CS1 maint: multiple names: authors list (link)
  3. ^ Durston AJ., Woltering JM (2008). "MiR-10 represses HoxB1a and HoxB3a in zebrafish". PLoS ONE. 3: e1396. PMID 18167555.
  4. ^ a b c Edwina McGlinn, Alysha Heimberg (2012). "Building a Robust A-P Axis". Current Genomics. 13: 278–288. doi:10.2174/138920212800793348. PMID 23204917.
  5. ^ a b c Richardson MK, Crooijmans RP, Groenen MA (2007). "Sequencing and genomic annotation of the chicken (Gallus gallus) Hox clusters, and mapping of evolutionarily conserved regions". Cytogenet Genome Res. 117 (1–4): 110–9. doi:10.1159/000103171. PMID 17675851.{{cite journal}}: CS1 maint: multiple names: authors list (link)
  6. ^ a b c d Tanzer A, Amemiya CT, Kim CB; et al. (2005). "Evolution of microRNAs located within Hox gene clusters". J Exp Zool B Mol Dev Evol. 304: 75–85. PMID 15643628. {{cite journal}}: Explicit use of et al. in: |author= (help); line feed character in |title= at position 47 (help)CS1 maint: multiple names: authors list (link)
  7. ^ a b c d Popovic R, Riesbeck LE, Velu CS; et al. (2009). "Regulation of mir-196b by MLL and its overexpression by MLL fusions contributes to immortalization". Blood. 113: 3314–22. doi:10.1159/000103171. PMID 19188669. {{cite journal}}: Explicit use of et al. in: |author= (help)CS1 maint: multiple names: authors list (link) Cite error: The named reference "pmid19188669" was defined multiple times with different content (see the help page).
  8. ^ Chopra VS, Mishra RK (2006). ""Mir"acles in hox gene regulation". Bioessays. 28 (5): 445–8. doi:10.1002/bies.20401. PMID 16615131.
  9. ^ a b Ronshaugen M, Biemar F, Piel J, Levine M, Lai EC (2005). "The Drosophila microRNA iab-4 causes a dominant homeotic transformation of halteres to wings". Genes Dev. 19 (24): 2947–52. doi:10.1101/gad.1372505. PMC 1315399. PMID 16357215.{{cite journal}}: CS1 maint: multiple names: authors list (link)
  10. ^ Chen C, Zhang Y, Zhang L, Weakley SM, Yao Q (2011). "MicroRNA-196: critical roles and clinical applications in development and cancer". J Cell Mol Med. 15 (1): 14–23. doi:10.1111/j.1582-4934.2010.01219.x. PMID 21091634.{{cite journal}}: CS1 maint: multiple names: authors list (link)
  11. ^ Mansfield JH, Harfe BD, Nissen R, Obenauer J, Srineel J, Chaudhuri A, Farzan-Kashani R, Zuker M, Pasquinelli AE, Ruvkun G, Sharp PA, Tabin CJ, McManus MT. (2004). "MicroRNA-responsive 'sensor' transgenes uncover Hox-like and other developmentally regulated patterns of vertebrate microRNA expression". Nat Genet. 36 (11): 1079–83. PMID 15361871.{{cite journal}}: CS1 maint: multiple names: authors list (link)
  12. ^ Yekta S, Shih IH, Bartel DP (2004). "MicroRNA-directed cleavage of HOXB8 mRNA". Science. 304 (5670): 594–6. doi:10.1126/science.1097434. PMID 15105502.{{cite journal}}: CS1 maint: multiple names: authors list (link)
  13. ^ a b c d Liu CJ, Tsai MM, Tu HF, Lui MT, Cheng HW, Lin SC. (2012). "miR-196a Overexpression and miR-196a2 Gene Polymorphism Are Prognostic Predictors of Oral Carcinomas". Ann Surg Oncol. PMID 23138850.{{cite journal}}: CS1 maint: multiple names: authors list (link)
  14. ^ Kawasaki H, Taira K (2004). "MicroRNA-196 inhibits HOXB8 expression in myeloid differentiation of HL60 cells". Nucleic Acids Symp Ser (Oxf) (48): 211–2. doi:10.1093/nass/48.1.211. PMID 17150553.
  15. ^ Maru DM, Singh RR, Hannah C; et al. (2009). "MicroRNA-196a is a potential marker of progression during Barrett's metaplasia-dysplasia-invasive adenocarcinoma sequence in esophagus". Am J Pathol. 174: 1940–8. PMID 19342367. {{cite journal}}: Explicit use of et al. in: |author= (help)CS1 maint: multiple names: authors list (link)
  16. ^ a b c d e f Matranga C, Tomari Y, Shin C; et al. (2005). "Passenger-strand cleavage facilitates assembly of siRNA into Ago2-containing RNAi enzyme complexes". Cell. 123 (20): 607. PMID 16271387. {{cite journal}}: Explicit use of et al. in: |author= (help)CS1 maint: multiple names: authors list (link) Cite error: The named reference "pmid16319892" was defined multiple times with different content (see the help page).
  17. ^ Xinjun Hea, Yi-Lin Yana, Johann K. Eberharta,b, Amaury Herpinc, Toni U. Wagnerc, Manfred Schartlc, and John H. Postlethwaita. (2011). "miR-196 regulates axial patterning and pectoral appendage initiation". Dev Biol. 357 (2): 463–477. PMID 21787766. {{cite journal}}: Text "doi:10.1016/j.ydbio.2011.07.014" ignored (help)CS1 maint: multiple names: authors list (link)
  18. ^ a b Sehm T, Sachse C, Frenzel C, Echeverri K (2009). "miR-196 is an essential early-stage regulator of tail regeneration, upstream of key spinal cord patterning events". Dev Biol. 334 (2): 468–80. doi:10.1016/j.ydbio.2009.08.008. PMID 19682983.{{cite journal}}: CS1 maint: multiple names: authors list (link)
  19. ^ Bloomston M, Frankel WL, Petrocca F; et al. (2007). "MicroRNA expression patterns to differentiate pancreatic adenocarcinoma from normal pancreas and chronic pancreatitis". JAMA. 297 (8): 1901. PMID 17473300. {{cite journal}}: Explicit use of et al. in: |author= (help)CS1 maint: multiple names: authors list (link)
  20. ^ Hui AB, Shi W, Boutros PC; et al. (2009). "Robust global micro-RNA profiling with formalin-fixed paraffin-embedded breast cancer tissues". Lab Invest.. 89: 597–606. PMID 19290006. {{cite journal}}: Explicit use of et al. in: |author= (help); line feed character in |title= at position 54 (help)CS1 maint: multiple names: authors list (link)
  21. ^ Woltering JM, Durston AJ (2008). "MiR-10 represses HoxB1a and HoxB3a in zebrafish". PLoS ONE. 3 (1): e1396. doi:10.1371/journal.pone.0001396. PMC 2148072. PMID 18167555.{{cite journal}}: CS1 maint: unflagged free DOI (link)
  22. ^ Velu CS, Baktula AM, Grimes HL. (2009). "Gfi1 regulates miR-21 and miR-196b to control myelopoiesis". Blood. 113: 4720. PMID 19278956.{{cite journal}}: CS1 maint: multiple names: authors list (link)
  23. ^ Akiko Shiotani , Noriya Uedo , Hiroyasu Iishi , Takahisa Murao ,Tomoko Kanzaki , Yoshiki Kimura , Tomoari Kamada ,Hiroaki Kusunoki , Kazuhiko Inoue and Ken Haruma. (2012). "H. pylori eradication did not improve dysregulation of specific oncogenic miRNAs in intestinal metaplastic glands". J Gastroenterol. 47: 988–998. PMID 22382634.{{cite journal}}: CS1 maint: multiple names: authors list (link)
  24. ^ a b Ohlsson Teague EM, Van der Hoek KH, Van der Hoek MB; et al. (2009). "MicroRNA-regulated pathways associated with endometriosis". Mol Endocrinol. 23 (75): 265. PMID 19074548. {{cite journal}}: Explicit use of et al. in: |author= (help)CS1 maint: multiple names: authors list (link)
  25. ^ Ye L, Wang X, Wang S; et al. (2009). "CD56+ T cells inhibit hepatitis C virus replication in human hepatocytes". Hepatology. 49 (62): 753. PMID 19085952. {{cite journal}}: Explicit use of et al. in: |author= (help)CS1 maint: multiple names: authors list (link)
  26. ^ Pedersen LM, Cheng G, Wieland S; et al. (2007). "Interferon modulation of cellular microRNAs as an antiviral mechanism". Nature. 449 (22): 919. PMID 17943132. {{cite journal}}: Explicit use of et al. in: |author= (help)CS1 maint: multiple names: authors list (link)
  27. ^ Sonkoly E, Stahle M, Pivarcsi A. (2008). "MicroRNAs and immunity: novel players in the regulation of normal immune function and inflammation". Semin Cancer Biol. 18 (40): 131. PMID 18291670.{{cite journal}}: CS1 maint: multiple names: authors list (link)
  28. ^ Kim YJ, Bae SW, Yu SS; et al. (2009). "miR-196a regulates proliferation and osteogenic differentiation in mesenchymal stem cells derived from human adipose tissue". J Bone Miner Res. 24 (25): 816. PMID 19063684. {{cite journal}}: Explicit use of et al. in: |author= (help)CS1 maint: multiple names: authors list (link)
  29. ^ Hou W, Tian Q, Zheng J, Bonkovsky HL (2010). "MicroRNA-196 represses Bach1 protein and hepatitis C virus gene expression in human hepatoma cells expressing hepatitis C viral proteins". Hepatology. 51 (5): 1494–504. doi:10.1002/hep.23401. PMC 2862129. PMID 20127796.{{cite journal}}: CS1 maint: multiple names: authors list (link)
  30. ^ Brest P, Lapaquette P, Mograbi B, Darfeuille-Michaud A, Hofman P. (2011). "Risk predisposition for Crohn disease". Autophagy. PMID 21508684.{{cite journal}}: CS1 maint: multiple names: authors list (link)

Further reading[edit]

  1. ^ Zhang XW, Pan SD, Feng YL, Liu JB, Dong J, Zhang YX, Chen JG, Hu ZB, Shen HB (2011). "[Relationship between genetic polymorphism in microRNAs precursor and genetic prediposition of hepatocellular carcinoma]". Zhonghua Yu Fang Yi Xue Za Zhi. 45 (3): 239–43. PMID 21624236.{{cite journal}}: CS1 maint: multiple names: authors list (link)
  2. ^ Georges M (2011). "The long and winding road from correlation to causation". Nat Genet. 43 (3): 180–1. doi:10.1038/ng0311-180. PMID 21350497.
  3. ^ Brest P, Lapaquette P, Souidi M, Lebrigand K, Cesaro A, Vouret-Craviari V, Mari B, Barbry P, Mosnier JF, Hébuterne X, Harel-Bellan A, Mograbi B, Darfeuille-Michaud A, Hofman P (2011). "A synonymous variant in IRGM alters a binding site for miR-196 and causes deregulation of IRGM-dependent xenophagy in Crohn's disease". Nat Genet. 43 (3): 242–5. doi:10.1038/ng.762. PMID 21278745.{{cite journal}}: CS1 maint: multiple names: authors list (link)
  4. ^ Chen C, Zhang Y, Zhang L, Weakley SM, Yao Q (2011). "MicroRNA-196: critical roles and clinical applications in development and cancer". J Cell Mol Med. 15 (1): 14–23. doi:10.1111/j.1582-4934.2010.01219.x. PMID 21091634.{{cite journal}}: CS1 maint: multiple names: authors list (link)
  5. ^ Scagnolari C, Zingariello P, Vecchiet J, Selvaggi C, Racciatti D, Taliani G, Riva E, Pizzigallo E, Antonelli G (2010). "Differential expression of interferon-induced microRNAs in patients with chronic hepatitis C virus infection treated with pegylated interferon alpha". Virol J. 7: 311. doi:10.1186/1743-422X-7-311. PMC 2996368. PMID 21070682.{{cite journal}}: CS1 maint: multiple names: authors list (link) CS1 maint: unflagged free DOI (link)
  6. ^ Li Y, Zhang M, Chen H, Dong Z, Ganapathy V, Thangaraju M, Huang S (2010). "Ratio of miR-196s to HOXC8 messenger RNA correlates with breast cancer cell migration and metastasis". Cancer Res. 70 (20): 7894–904. doi:10.1158/0008-5472.CAN-10-1675. PMC 2955846. PMID 20736365.{{cite journal}}: CS1 maint: multiple names: authors list (link)
  7. ^ Guan Y, Mizoguchi M, Yoshimoto K, Hata N, Shono T, Suzuki SO, Araki Y, Kuga D, Nakamizo A, Amano T, Ma X, Hayashi K, Sasaki T (2010). "MiRNA-196 is upregulated in glioblastoma but not in anaplastic astrocytoma and has prognostic significance". Clin Cancer Res. 16 (16): 4289–97. doi:10.1158/1078-0432.CCR-10-0207. PMID 20601442.{{cite journal}}: CS1 maint: multiple names: authors list (link)
  8. ^ Asli NS, Kessel M (2010). "Spatiotemporally restricted regulation of generic motor neuron programs by miR-196-mediated repression of Hoxb8". Dev Biol. 344 (2): 857–68. doi:10.1016/j.ydbio.2010.06.003. PMID 20553899.
  9. ^ Schotte D, Lange-Turenhout EA, Stumpel DJ, Stam RW, Buijs-Gladdines JG, Meijerink JP, Pieters R, Den Boer ML (2010). "Expression of miR-196b is not exclusively MLL-driven but is especially linked to activation of HOXA genes in pediatric acute lymphoblastic leukemia". Haematologica. 95 (10): 1675–82. doi:10.3324/haematol.2010.023481. PMC 2948092. PMID 20494936.{{cite journal}}: CS1 maint: multiple names: authors list (link)
  10. ^ Hou W, Tian Q, Zheng J, Bonkovsky HL (2010). "MicroRNA-196 represses Bach1 protein and hepatitis C virus gene expression in human hepatoma cells expressing hepatitis C viral proteins". Hepatology. 51 (5): 1494–504. doi:10.1002/hep.23401. PMC 2862129. PMID 20127796.{{cite journal}}: CS1 maint: multiple names: authors list (link)
  11. ^ McGlinn E, Yekta S, Mansfield JH, Soutschek J, Bartel DP, Tabin CJ (2009). "In ovo application of antagomiRs indicates a role for miR-196 in patterning the chick axial skeleton through Hox gene regulation". Proc Natl Acad Sci U S A. 106 (44): 18610–5. doi:10.1073/pnas.0910374106. PMC 2773993. PMID 19846767.{{cite journal}}: CS1 maint: multiple names: authors list (link)
  12. ^ Sehm T, Sachse C, Frenzel C, Echeverri K (2009). "miR-196 is an essential early-stage regulator of tail regeneration, upstream of key spinal cord patterning events". Dev Biol. 334 (2): 468–80. doi:10.1016/j.ydbio.2009.08.008. PMID 19682983.{{cite journal}}: CS1 maint: multiple names: authors list (link)
  13. ^ Popovic R, Riesbeck LE, Velu CS, Chaubey A, Zhang J, Achille NJ, Erfurth FE, Eaton K, Lu J, Grimes HL, Chen J, Rowley JD, Zeleznik-Le NJ (2009). "Regulation of mir-196b by MLL and its overexpression by MLL fusions contributes to immortalization". Blood. 113 (14): 3314–22. doi:10.1182/blood-2008-04-154310. PMC 2665896. PMID 19188669.{{cite journal}}: CS1 maint: multiple names: authors list (link)
  14. ^ Sonkoly E, Ståhle M, Pivarcsi A (2008). "MicroRNAs and immunity: novel players in the regulation of normal immune function and inflammation". Semin Cancer Biol. 18 (2): 131–40. doi:10.1016/j.semcancer.2008.01.005. PMID 18291670.{{cite journal}}: CS1 maint: multiple names: authors list (link)
  15. ^ Woltering JM, Durston AJ (2008). "MiR-10 represses HoxB1a and HoxB3a in zebrafish". PLoS ONE. 3 (1): e1396. doi:10.1371/journal.pone.0001396. PMC 2148072. PMID 18167555.{{cite journal}}: CS1 maint: unflagged free DOI (link)
  16. ^ Richardson MK, Crooijmans RP, Groenen MA (2007). "Sequencing and genomic annotation of the chicken (Gallus gallus) Hox clusters, and mapping of evolutionarily conserved regions". Cytogenet Genome Res. 117 (1–4): 110–9. doi:10.1159/000103171. PMID 17675851.{{cite journal}}: CS1 maint: multiple names: authors list (link)
  17. ^ Kawasaki H, Taira K (2004). "MicroRNA-196 inhibits HOXB8 expression in myeloid differentiation of HL60 cells". Nucleic Acids Symp Ser (Oxf) (48): 211–2. doi:10.1093/nass/48.1.211. PMID 17150553.
  18. ^ Chopra VS, Mishra RK (2006). ""Mir"acles in hox gene regulation". Bioessays. 28 (5): 445–8. doi:10.1002/bies.20401. PMID 16615131.
  19. ^ Ronshaugen M, Biemar F, Piel J, Levine M, Lai EC (2005). "The Drosophila microRNA iab-4 causes a dominant homeotic transformation of halteres to wings". Genes Dev. 19 (24): 2947–52. doi:10.1101/gad.1372505. PMC 1315399. PMID 16357215.{{cite journal}}: CS1 maint: multiple names: authors list (link)
  20. ^ Hornstein E, Mansfield JH, Yekta S, Hu JK, Harfe BD, McManus MT, Baskerville S, Bartel DP, Tabin CJ (2005). "The microRNA miR-196 acts upstream of Hoxb8 and Shh in limb development". Nature. 438 (7068): 671–4. doi:10.1038/nature04138. PMID 16319892.{{cite journal}}: CS1 maint: multiple names: authors list (link)
  21. ^ Yekta S, Shih IH, Bartel DP (2004). "MicroRNA-directed cleavage of HOXB8 mRNA". Science. 304 (5670): 594–6. doi:10.1126/science.1097434. PMID 15105502.{{cite journal}}: CS1 maint: multiple names: authors list (link)
  22. ^ Mansfield JH, Harfe BD, Nissen R, Obenauer J, Srineel J, Chaudhuri A, Farzan-Kashani R, Zuker M, Pasquinelli AE, Ruvkun G, Sharp PA, Tabin CJ, McManus MT. (2004). "MicroRNA-responsive 'sensor' transgenes uncover Hox-like and other developmentally regulated patterns of vertebrate microRNA expression". Nat Genet. 36 (11): 1079–83. PMID 15361871.{{cite journal}}: CS1 maint: multiple names: authors list (link)
  23. ^ Chen F, Capecchi MR. (1997). "Targeted mutations in hoxa-9 and hoxb-9 reveal synergistic interactions". Dev Biol. 181 (2): 186-96. PMID 9013929.

External links[edit]



Category:MicroRNA