Saturday, December 1, 2012

MITF and Melanoma; Targeting Specific Pathway Elements

MITF is a transcription factor which has been linked to melanoma. It can be activated in the RAF/BRAF pathway and thus when activated results in the loss of apoptosis and the migration of cells.

What is MITF?

As noted by Carreira et al[1]:

It is widely held that cells with metastatic properties such as invasiveness and expression of matrix metalloproteinases arise through the stepwise accumulation of genetic lesions arising from genetic instability and “clonal evolution.”

By contrast, we show here that in melanomas invasiveness can be regulated epigenetically by the microphthalmia-associated transcription factor, Mitf, via regulation of the DIAPH1 gene encoding the diaphanous-related formin Dia1 that promotes actin polymerization and coordinates the actin cytoskeleton and microtubule networks at the cell periphery.

Low Mitf levels lead to down-regulation of Dia1, reorganization of the actin cytoskeleton, and increased ROCK-dependent invasiveness, whereas increased Mitf expression leads to decreased invasiveness.

Significantly the regulation of Dia1 by Mitf also controls p27Kip1-degradation such that reduced Mitf levels lead to a p27Kip1-dependent G1 arrest. Thus Mitf, via regulation of Dia1, can both inhibit invasiveness and promote proliferation.

The results imply variations in the repertoire of environmental cues that determine Mitf activity will dictate the differentiation, proliferative, and invasive/migratory potential of melanoma cells through a dynamic epigenetic mechanism.

Note that the discussion above shows that overexpression of MITF leads to less invasiveness. We show the details of the pathway dynamics below.
 They continue:

Although Mitf is clearly required for melanoma proliferation, why it is necessary has not been previously established. To understand how Mitf depletion led to a block in G1/S transition, we examined a number of known markers of proliferation and the cell cycle. Western blotting of cells transfected with control or Mitfspecific siRNA  revealed that depletion of Mitf led, as expected from our previous work, to decreased expression of p21Cip1, but intriguingly also induced expression of the p27Kip1 cyclin-dependent kinase inhibitor. Note that similar results were obtained using a second Mitf siRNA directed against a different region of Mitf. We also observed reduced expression of cyclin E, and PCNA, most likely as an indirect effect of the cell cycle arrest, but no change in cyclin D1 or Cdk2 levels. Tubulin and lamin B were used as loading controls.

Finally they note:

In summary, Mitf appears to lie right at the heart of the melanocyte, coordinating survival, cell cycle entry and exit, cytoskeletal organization, melanosome assembly and transport, differentiation and migration/metastasis. As such, understanding Mitf regulation and function may well be the key to achieving one of the major aims of cancer research, an effective melanoma therapy.

Thus the MITF function, in their view, is critical.

MITF Targeting and Control

From a current Eureka posting, Zoufal writes of work recently reported in Genes & Development by Pogenberg[2] et al[3] focused upon targeting and thus controlling MITF:

The results, published in the scientific journal "Genes & Development", throw new light on the workings of the so-called Microphthalmia-associated Transcription Factor MITF, that is not only connected to skin cancer, but also to a variety of hereditary diseases where the production of the skin pigment melanin is disturbed, and to certain aspects of ageing. "Our data could provide a rational basis for the development of tailor-made drugs targeting MITF", explains first author Vivian Pogenberg from the Hamburg branch of the European Molecular Biology Laboratory

But MITF also makes stem cells turn into melanocytes in the first place and controls cell proliferation and death in these cells. That's why MITF is called a master regulator. In fact, it also has functions in other cell types like mast cells of the immune system and bone eating osteoclasts...

Mutations in MITF not only play a role in the development of skin cancer, but also cause severe genetic diseases like the Tietz and Waardenburg syndromes that lead to deafness, skin and hair pigmentation defects, abnormal eye anatomy and altered vision. The transcription factor also plays a role in our hair turning grey with age and other age-related pigmentation alterations…

Crystals scatter X-rays in characteristic ways and produce diffraction patterns from which the structure of the crystal - and here MITF - can be reconstructed. The analysis revealed unexpected molecular insertions that give MITF a unique kink. MITF forms a dimer with a long coiled-coil protein "zipper", and the kink in this zipper limits MITF's ability to bind to other transcription factors.

This paper thus appears to provide a targeted means to control MITF and then perhaps melanoma metastasis. Identification of binding sites is essential to control.

More on MITF

We can provide a bit more history on MITF and its functions. As NCBI states[4], MITF, microphthalmia-associated transcription factor (3p14.2-p14.1):

This gene encodes a transcription factor that contains both basic helix-loop-helix and leucine zipper structural features. It regulates the differentiation and development of melanocytes retinal pigment epithelium and is also responsible for pigment cell-specific transcription of the melanogenesis enzyme genes. Heterozygous mutations in the this gene cause auditory-pigmentary syndromes, such as Waardenburg syndrome type 2 and Tietz syndrome

MITF has been known to be a key player in melanoma metastasis. As Chin et al state in their review paper[5]:

MITF, a melanoma oncogene targeted by amplification

The promise of DNA-based structural alterations as the entry point for gene discovery has been illustrated by the recent identification of MITF as a melanoma oncogene. The discovery of MITF amplification in melanoma derived from an integrated analysis of genomic copy gains and losses, together with sample-matched mRNA expression data.

When clustering algorithms were applied to SNP array-derived chromosomal copy number data generated for the NCI-60 cancer cell line collection, some of these cell lines aggregated according to tissue of origin, including several melanoma cell lines. The bidimensionality of the hierarchical algorithm also enabled the identification of chromosomal alterations driving these lineage-restricted clustering patterns, and suggested that lineage-specific cancer genes might reside within the genomic regions implicated.

For the melanoma cell lines, the common genomic alteration was a region of copy gain at chromosome 3p14-3p13. To facilitate the identification of an oncogene targeted by this amplification event, the NCI- 60 collection was partitioned based on the presence or absence of copy gain at the relevant chromosome 3p locus. This partitioning served as a two-class distinction that drove a supervised analysis of sample-matched gene expression data. Although the gene expression signature that emerged was dominated by melanocyte lineage genes (as expected given that only melanoma cell lines comprised the 3p-amplified class), MITF was the only gene showing significantly increased expression in association with the 3p-amplified melanoma cell lines that also mapped to the common region of 3p copy gain.

MITF amplification was subsequently detected in 10% of primary cutaneous and 15%–20% of metastatic melanomas.

Although the majority of amplifications were low level (e.g., four to six copies per cell), high-level amplicons were also observed, including one sample that exhibited >100 copies per diploid genome. A Kaplan- Meier analysis performed on metastatic melanomas suggested that MITF amplification in this setting correlated with adverse 5-yr patient survival.

Finally, ectopic MITF overexpression complemented BRAFV600E in conferring soft agar colony growth to immortalized melanocytes engineered to express TERT, and to lack the pRB and p53 pathways. These functional studies thereby suggested a genetic context that might characterize a subset of human melanomas whose survival is dependent on MITF.

MITF also exemplifies a newly recognized “lineage survival” oncogenic mechanism, wherein tumor genetic alterations may target survival functions also operant in the relevant cellular lineages during development and differentiation.

Thus, while the discovery of MITF amplification began as a systematic genomics-based survey of many human cancer types, it provides a striking convergence of melanoma oncogene discovery and melanocyte development.

Next, as Genovese et al state[6]:

Histidine triad nucleotide-binding protein 1 (HINT1) is a haploinsufficient tumor suppressor gene that inhibits the Wnt/β-catenin pathway in colon cancer cells and Microphthalmia-associated transcription factor (MITF) activity in human mast cells. MITF and β-catenin play a central role in melanocyte and melanoma cell survival, and this study aimed to investigate the effects of HINT1 on the MITF and β-catenin pathways in malignant melanoma cells.

We found that HINT1 inhibits MITF and β-catenin transcriptional activity, and both proteins can be co-immunoprecipitated with an anti-HINT1-specific antibody in melanoma cell lines. Stable, constitutive overexpression of the HINT1 protein in human melanoma cells significantly impaired cell proliferation in vitro and tumorigenesis in vivo.

These effects were associated with a decreased expression of cyclin D1 and BCL2, well known MITF and β-catenin transcription targets, respectively. We also demonstrated that BCL2 and cyclin D1 can partially rescue the HINT1-driven phenotype. Moreover, we found in ChIP assays that HINT1 binds the chromatin at MITF and β-catenin sites in BCL2 and cyclin D1 promoters, respectively, and that mSIN3a and HDAC1, well known transcriptional repressors, can be co-immunoprecipitated with an anti-HINT1-specific antibody. These findings support the tumor suppressor activity of HINT1 gene in melanoma cells by promoting the formation of non-functional complexes with oncogenic transcription factors like MITF and β-catenin.

Finally as Larribere et al state[7]:

Microphthalmia-associated transcription factor (MITF) M-form is a melanocyte-specific transcription factor that plays a key role in melanocyte development, survival, and differentiation. Here, we identified MITF as a new substrate of caspases and we characterized the cleavage site after Asp 345 in the C-terminal domain.

We show that expression of a non-cleavable form of MITF renders melanoma cells resistant to apoptotic stimuli, and we found that the C-terminal fragment generated upon caspase cleavage is endowed with a proapoptotic activity that sensitizes melanoma cells to death signals. The proapoptotic function gained by MITF following its processing by caspases provides a tissue-restricted means to modulate death in melanocyte and melanoma cells.

Their observation does show the impact of MITF in the control of melanoma.

Observations

The recent work demonstrates that we can through an understanding of the pathways then target specific pathway control proteins by understanding their structure. We can already control B RAF in certain circumstances by targeting its specificity and that controlling the path but allowing MITF control in a broad sense may actually be much more powerful if the results hold for clinical applications.

The ability to find, characterize, and design binding site specific blocking agents is an essential step in a broader control of multiple cancers.


[1] Carreira, S., et al, Mitf regulation of Dia1 controls melanoma proliferation and invasiveness, Genes Dev. 2006 20: 3426-3439.

[2] See Pogenberg in http://genesdev.cshlp.org/

[3] Zoufal, T., X-ray analysis deciphers master regulator important for skin cancer, http://www.eurekalert.org/pub_releases/2012-12/ded-xad113012.php

[5] Chin et al, Malignant melanoma: genetics and therapeutics in the genomic era, Genes Dev. 2006 20: 2149-2182

[6] Genovese G, et al, The tumor suppressor HINT1 regulates MITF and β-catenin transcriptional activity in melanoma cells, Cell Cycle. 2012 Jun 1; 11(11):2206-15.

[7] Larribere, et al, The cleavage of microphthalmia-associated transcription factor, MITF, by caspases plays an essential role in melanocyte and melanoma cell apoptosis, Genes Dev. 2005 19: 1980-1985.