Next Article in Journal
GIRK Channels as Candidate Targets for the Treatment of Substance Use Disorders
Previous Article in Journal
Chronic Endothelial Dysfunction after COVID-19 Infection Shown by Transcranial Color-Coded Doppler: A Cross-Sectional Study
Previous Article in Special Issue
Long-Term Response to Tyrosine Kinase Inhibitors for Metastatic Renal Cell Carcinoma
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Article

The Tumour Suppressor Fhit Protein Activates C-Raf Ubiquitination and Degradation in Human Melanoma Cells by Interacting with Hsp90

by
Francesco Paduano
1,2,*,†,
Eugenio Gaudio
3,*,† and
Francesco Trapasso
1
1
Department of Experimental and Clinical Medicine, University Magna Græcia of Catanzaro, 88100 Catanzaro, Italy
2
Stem Cells and Medical Genetics Units, Tecnologica Research Institute and Marrelli Health, 88900 Crotone, Italy
3
DTI-Tech, 6500 Bellinzona, Switzerland
*
Authors to whom correspondence should be addressed.
These authors contributed equally to this work.
Biomedicines 2022, 10(10), 2551; https://doi.org/10.3390/biomedicines10102551
Submission received: 1 September 2022 / Revised: 10 October 2022 / Accepted: 11 October 2022 / Published: 13 October 2022
(This article belongs to the Special Issue Feature Papers in Cancer Biology and Therapeutics)

Abstract

:
Fhit protein expression is reduced in the majority of human tumors; moreover, its restoration both triggers apoptosis of cancer cells and suppresses tumor formation in a large number of preclinical models of cancers. In the following study, we observed that Fhit expression is significantly reduced in human melanoma cells, and their in vivo growth is blocked by a recombinant adenovirus carrying the FHIT gene. Importantly, we found here that Fhit physically interacts with Hsp90. Since Hsp90 is a chaperone with a crucial function in the conformational maturation and stabilization of C-Raf, we also investigated whether Fhit could interfere with the Hsp90/C-Raf protein complex in melanoma. Interestingly, the administration of the Hsp90 inhibitor 17-AAG, in combination with Fhit protein overexpression in melanoma cells, reacts synergistically to increase C-Raf ubiquitination and degradation. These data reveal Hsp90 as a novel interactor of Fhit and suggest that FHIT activity restoration could represent a helpful strategy for suppressing the oncogenic C-Raf pathway in the therapy of human melanoma.

1. Introduction

The FHIT gene maps on chromosome 3p14.2 and encompasses the most active fragile site in the human genome, known as FRA3B. The t(3;8) (p14.2-q24) translocation, which leads to impaired Fhit protein expression, was discovered in 1996 in a family affected by renal cancer [1]. Although early in vitro studies showed that Fhit is an enzyme able to hydrolyse diadenosine triphosphates, which are produced in cells in response to stress [2], its activity as a tumor-suppressing protein—although frequently assessed both in experimental and human tumorigenesis [3,4]—remained obscure for a long time since no protein partners were proven for a long time after its discovery. Only more than a decade after its identification has our group identified a list of Fhit candidate partners, including among them, FRDX, HSP10 [4], and annexin A4 (A4) [5]. We also identified a short 7-aminoacids peptide from the Fhit protein that is still able to bind A4 and sensitize lung cancer cells to paclitaxel, thus recapitulating the activity of the full-length Fhit protein on A4 [6,7].
Even though some investigations reported that FHIT hypermethylation, a common mechanism of FHIT inactivation, in uveal melanoma is a very rare event [8,9], the role of Fhit protein in human melanogenesis was never deeply investigated.
Melanoma is considered one of the most aggressive human cancers and one of the most difficult cancers to treat due to its resistance to current therapies [10,11]. Despite the advances in metastatic melanoma treatment, including target therapy and immunotherapy, this disease is very problematic to treat and still remains a lethal type of cancer [12,13]. For example, it has been shown that 5-year survival after diagnosis for metastatic melanoma is around 50% when treated with combination immunotherapy and 33% with combination BRAF/MEK targeted therapy [13].
Melanoma harbors multiple genetic alterations that corrupt biological pathways controlling cell growth, differentiation, survival, and metabolic reprogramming [11,14]. Among these pathways, the MAPK cascade, including RAS, RAF, MEK, and ERK, as well as other signaling pathways, such as WNT and PI3K-AKT, have been shown to be frequently mutated in melanoma [11,14]. Genetic alterations in BRAF and NRAS genes are very common in primary melanomas, whereas other mutations are involved in NRAS, TP53, TERT, CDKN2A, and PTEN genes [15]. For example, the BRAF V600E is the most frequent genetic cause of melanoma, which is responsible for the increased proliferation and metabolic reprogramming of melanoma cells [11,16].
Although today there are several successful melanoma therapies, such as approaches that primarily suppress the BRAF oncoprotein pathway [13], it is very important to investigate new therapeutic approaches and molecular targets. This happens because it has been shown that 15–20% of primary melanomas can become resistant to drugs, such as BRAF/MEK inhibitors [14,15].
Here, we demonstrated that Fhit negatively modulates C-Raf expression in melanoma cells by interacting with Hsp90. Moreover, the restoration of Fhit expression in Fhit-negative melanoma cells triggers apoptosis and blocks tumor formation in vivo. These results suggest a further pathway to be explored for the generation of novel tools for the therapeutic targeting of human melanoma.

2. Materials and Methods

2.1. Transfection and Cell Cultures

Cell cultures and experiments with transfections were executed as previously described [5]. Briefly, melanoma cell lines were cultivated at 37 °C and 5% CO2. HEK293 cells were used for the construction and amplification of the recombinant adenovirus vectors.

2.2. Immunoblotting Analysis

Immunoblotting analyses were performed as previously described [17,18,19,20]. Briefly, enriched plasma membrane proteins were carried out using a specific kit (Bio-Rad, Pleasanton, CA, USA). These proteins were then analyzed on polyacrylamide gels and subsequently moved to membranes of nitrocellulose (Bio-Rad). Subsequently, blocked membranes were incubated with primary antibodies and detected by the specific secondary antibodies. Co-IP experiments were carried out as previously described [20]. Briefly, total lysates were incubated with the anti-Hsp90 antibody at 4 °C O.N. Subsequently, antibodies or beads were washed and boiled in a buffer containing SDS, and then proteins were analyzed on polyacrylamide gels (4–20%) with or without the use of the crosslinking agent known as [(dithiobis(succinimidylpropionate), or DSP]).

2.3. Protein Interaction Evaluation

Co-immunoprecipitation (Co-IP) assays were performed as previously described [20]. Briefly, enriched plasma membrane proteins, with or without the addition of DSP, were incubated with magnetic agarose beads (NIN-TA, Qiagen, Washington, USA), and the resulting interacting proteins were obtained.

2.4. Construction of Recombinant Adenoviral Vectors

AdFHIT (an adenovirus carrying the FHIT cDNA) and AdFHIT-His6 (an adenovirus carrying the FHIT cDNA with a His6 epitope tag) were obtained as previously described [5]. Detailed methods are described in the Supplementary Materials and Methods.

2.5. In-Vitro Cell Growth Evaluation

Melanoma cells, seeded on plates, were infected with an adenovirus carrying FHIT cDNA (AdFHIT), GFP cDNA (AdGFP), or a control (mock-infected). Subsequently, cells were counted at 24 h intervals.

2.6. Proteomic Experiments

Mass spectrometry studies, as well as a digestion and MALDI analysis, LTQ, and protein identification, were carried out as previously described [21]. Detailed methods are described in the Supplementary Materials.

2.7. TUNEL Assay

Tunel assays were performed as previously described [5]. Briefly, apoptosis of the A549 cells was measured by a TUNEL assay (Boehringer/Roche, Indianapolis, IN, USA).

2.8. Ubiquitination Assays

Protein lysates were incubated with O.N. with an antibody against C-Raf (Cell Signaling, Beverly, MA, USA) or AG/agarose beads and subsequently separated on polyacrylamide gels (4–20%). Finally, obtained membranes of nitrocellulose were immunoblotted with an antibody against HA-HRP to detect Ub-HA (ubiquitin) and antibodies against Fhit and C-Raf (Cell Signaling, Beverly, MA, USA).

2.9. Immunohistochemistry

Colo38 cells on the slide were hydrated in PBS for 5 min. Next, slides were soaked in TBS with tween for 5 min, quenched with 0.03% H2O2 for 5 min, and then rinsed with TBST. The primary antibody, C-Raf (LifeSpan BioSciences Seattle, WA, USA), was diluted at 1:1000 and then incubated for 1 h at RT. The color reaction was developed with Red Vulcan for 10 min. Lastly, slides were stained with Mayer’s Haematoxylin and mounted.

2.10. Animal Studies

Animal studies were carried out under the guidelines validated by the animal facility of Ohio State University. Tumors were evaluated as previously described [5]. Briefly, Colo38 cells were injected into the right flanks of female nude mice (nu/nu) at 6 weeks of age. The following formula estimated tumor volumes: V (in mm3) = A × B2/2, in which A was the measure of the largest diameter and B was the measure of the perpendicular diameter.

2.11. Statistics

Results are expressed as the mean ± standard deviation of three separate experiments. The significance of the differences between groups was evaluated using Student’s t-tests or one-way ANOVAs. Analysis was conducted using GraphPad Prism software (San Diego, CA, USA), and differences were considered significant if p < 0.05.
Values were represented as means ± SD derived from 3 independent experiments. Differences among groups were calculated using Student’s t-tests, and significance was determined at p < 0.05.

3. Results

3.1. Identification of Hsp90 as a Candidate Fhit Protein Interactor

As described in our previously published study, Hsp90 was identified through mass spectrometry as a putative partner of the Fhit protein [5]. We analyzed the physical interaction between these two proteins as described below.

3.2. Fhit Interacts with Hsp90

To determine whether Fhit and Hsp90 physically interacted, we performed a co-immunoprecipitation experiment. As shown in Figure 1A, Colo38 melanoma cells were infected with Ad-FHIT and subsequently treated with DSP. The resultant proteins were immunoprecipitated with protein A/G agarose and IgG or anti-Hsp90 antibodies and, finally, were tested using Western blotting. These results confirmed that the Fhit protein interacts with Hsp90 (Figure 1A). Similarly, the interaction between Fhit and Hsp90 was further confirmed through immunoprecipitation experiments, where cells were infected with an adenovirus carrying FHIT cDNA modified with a sequence encoding a His6 epitope tag (AdFHIT-His6) at its 3′ end (data not shown).
We subsequently analyzed Fhit protein expression in several melanoma cell lines, including SK-Mel28, FO-1, E6LCP, 1259CL93, M14, Hs294T, 1106, and Colo38 having mutations in B-RAF and WM13066, MeWo and CHL-1 having B-RAF WT, as well as normal melanocytes via Western blotting (Figure 1B and data not shown). Results indicated that the levels of Fhit expression were lower in some melanoma cell lines with respect to their normal counterpart.

3.3. Fhit Affects C-Raf Expression in Human Melanoma

Since it is well known that Hsp90 regulates C-Raf folding, we sought to determine whether Fhit affected C-Raf expression through Hsp90 via Western blotting. To this end, melanoma cell lines, Colo38 (B-RAF V600E) and CHL-1 (B-RAF wild-type), were mock- and FHIT-transfected and treated with the inhibitor of Hsp90 known as 17-allylamino-17-dimethoxy geldanamycin (17-AAG) for 24 hrs (Figure 2A,B). Results showed that the C-Raf and p-ERK expressions were downregulated through Fhit expression (Figure 2A,B; lane 4 vs. lane 1) and 17-AAG treatment (lanes 5, 6 vs. lanes 2, 3). Hsp90 weakly regulates B-Raf expression, and for this reason, 17-AAG does not affect much B-Raf expression. It is probable that B-Raf expression is increased as a compensatory effect of C-Raf downregulation. Significantly, since an exogenous expression of Fhit was responsible for the reduced expression of C-Raf, we performed a ubiquitination assay. To this end, Colo38 cells were transfected with Ub-HA and FHIT expression vectors. After 48 h of transfection, cells were exposed to 17-AAG and the proteasome inhibitor, MG132, at a concentration of 10 μM for an additional three hours. Subsequently, after the lysis of the cells, the resultant proteins were immunoprecipitated against C-Raf with an antibody, followed by immunoblotting with an antibody against the HA epitope. Results revealed that the expression of Fhit, combined with additional treatment with 17-AAG, resulted in C-Raf ubiquitination (data not shown). These data suggest that Fhit expression disturbs Hsp90’s activity and enhances 17-AAG effects by producing, as the read-out, the severe ubiquitination of C-Raf. Significantly, melanoma cells shift their dependency from B-Raf to C-Raf and vice versa; we were not able to see any regulation or ubiquitination of B-Raf by Fhit alone or in combination with 17-AAG.

3.4. Restoration of Fhit Expression Affects Proliferation of Melanoma Cells

To determine whether the FHIT gene could be used as a therapeutic agent, we constructed a replication-defective adenovirus encompassing the FHIT gene and a GFP reporter gene as previously described [22]. Colo38 and CHL-1 cells were infected with AdFHIT or AdGFP, and levels of adenovirus infection were observed using confocal microscopy 48–72 h after infection. Subsequently, exogenous Fhit protein expression was evaluated through Western blotting. Data showed that the restoration of Fhit expression significantly affected the proliferation of melanoma cells, such as Colo38 and CHL-1 (Figure 3A).

3.5. Restoration of Fhit Induces Apoptosis in Human Melanoma

We also examined the role of Fhit expression on the proliferation of E6LCP and Colo38 melanoma cells through MTS. Interestingly, Colo38 melanoma cells, which do not express Fhit endogenously, were strongly affected, in terms of proliferation, by the re-expression of adenovirus-mediated Fhit. On the contrary, E6LCP melanoma cells expressing Fhit endogenously were slightly affected by an over-expression of Fhit (Figure 3A). In addition, Colo38 melanoma cells were analyzed using TUNEL assays to evaluate the apoptotic effects of Fhit gene therapy alone or combined with 17-AAG (Figure 3B). Results showed that the exogenous expression of Fhit induces apoptosis in human melanoma Colo38, and that the levels of apoptosis were further increased when using 17-AAG on both days 1 and 2 after treatment.
To corroborate the effect of AdFHIT on melanoma cell proliferation, we treated Colo38 with AdFHIT and AdGFP, alone or in combination with 17-AAG (Figure 3C).
Results showed that the proliferation of melanoma AdFHIT-infected cells was significantly reduced after 3 days of treatment. Importantly, Colo38 proliferation levels were further decreased when using 17-AAG in combination with AdFHIT (Figure 3D).

3.6. Fhit Regulates C-Raf Expression in Melanoma Cells

Subsequently, C-Raf protein expressions were assessed by immunohistochemistry on mock- and Fhit-transfected Colo38 melanoma cells (Figure 4A). Immunohistochemistry assays demonstrated a significant reduction in C-Raf protein expression when Fhit was overexpressed by transfection, which confirmed our Western blotting data.
Using mock- and FHIT-transfected Colo38 melanoma cells, subsequently treated with DMSO, Sorafenib (an inhibitor of C-Raf and mutated B-Raf), and PLX4032 (an inhibitor of mutated B-Raf), we observed the inactivating phosphorylation of C-Raf on Ser 289/296/301 was relevant in all FHIT-transfected conditions. An additive effect between Fhit and the kinase inhibitor drug treatments was detected (Figure 4B).

3.7. Tumour Suppressor Activity of Fhit in Melanoma Xenograft

Finally, we checked the effects of Fhit in vivo in a preclinical model of melanoma. To this end, Colo38 cells pre-infected with AdFHIT at MOI 50 were injected into the flank of nude mice. After ten days of injection, tumors of around 8 mm in diameter were observed in some groups of mice. Results showed that tumors appeared both in the control groups of mice injected with mock-infected cells or AdGFP-infected cells. Importantly, xenograft tumors treated with AdFHIT displayed a significant reduction in volume and weight with respect to the control group (Figure 5A,B).

4. Discussion

In this study, by using adenovirus vectors and a proteomic approach, we identified Hsp90 as a novel interactor of Fhit. The methodological approach used in this study was previously successfully used to isolate and characterize soluble Fhit protein complexes, including Hsp10/Hsp60, ferredoxin reductase [4], and annexin 4 [5]. Since it is well known that Fhit is downregulated in most cancer cell lines [1,2], we also extended this investigation to melanoma cell lines, where it appears mostly downregulated. Moreover, to clarify Fhit tumor suppressor activity in melanoma, we studied Fhit protein complexes using our validated proteomic approach [5,21].
In this study, Hsp90 was recognized as a candidate Fhit partner, and co-immunoprecipitation experiments confirmed their interaction. Importantly, Hsp90 is crucial for the folding of many oncogenic proteins [23]. Hsp90 is a chaperone, playing a crucial function in the folding and stabilizing of many signaling oncoproteins, and is essential for the correct folding of C-Raf [23,24,25,26]. It was shown that the inhibition of the heat shock protein 90 (Hsp90) function with geldanamycin, or its derived 17-allylamino-17-demethoxygeldanamycin (17-AAG), effectively causes the ubiquitination of C-Raf in the proteasome [27]. Significantly, the folding of Raf family members, such as A-Raf, B-Raf, and C-Raf, is mediated by Hsp90 and other players, such as cdc37, Hop, and Hsp70. These complexes fold and stabilize several proteins involved in cancer pathogenesis [28]. C-Raf and its related RAS/RAF/MEK pathway are hyperactivated in 30% of human cancers and most melanomas. Notably, the B-Raf gene was observed to be mutated in 50% of human melanomas. A strong proliferation and survival rate characterized the cancer cells with the V600E B-Raf allele (B-Raf V600E), and these cells are also more sensitive to 17-AAG than wild-type protein [29].
Since C-Raf is a kinase overexpressed in melanoma, and its folding is under Hsp90 control [30], we studied the role of Fhit in this complex. Here, we observed that the direct interaction between Fhit and Hsp90 can affect Hsp90 activity and, subsequently, C-Raf expression. We demonstrated that C-Raf was ubiquitinated under the combined treatment of Fhit expression and 17-AAG much more than under a single treatment.
Melanoma is one of the most aggressive human tumors, and its incidence and mortality rate have increased worldwide in the last five years [31]. We measured the protein expression levels of Fhit in a panel of twelve melanoma human cell lines and demonstrated that Fhit expression in such cells was reduced or undetectable compared to primary normal human melanocytes. Immunohistochemistry assays showed a significant reduction in C-Raf in Colo38 cells when Fhit was overexpressed by transfection.
To assess FHIT activity as a therapeutic gene in melanoma, we generated a recombinant adenovirus encompassing the human cDNA of FHIT (AdFHIT). Our experiments established that Fhit has therapeutic activity, both in vitro and in vivo, on melanoma cells. Fhit-negative Colo38 melanoma cells transduced with AdFHIT showed a significantly reduced proliferation rate and underwent programmed cell death. The in vivo investigation of AdFHIT as a therapeutic gene in melanoma was further assessed on a melanoma preclinical xenograft model established by subcutaneously injecting human melanoma cells in nude mice. In vivo, FHIT expression effectively reduced tumor growth compared to the control.
Our research group has a long history of studies on the role of Fhit in tumorigenesis [7,32] and, more recently, on the identification of Fhit partners [4,5,6]. All identified Fhit interactors partly shared among different types of cancer cell lines, including lung cancer and melanoma, were investigated in order to both uncover their functional role in cancer and to try to propose anticancer solutions with novel mechanisms of action. In this study, our data suggest that the re-expression of Fhit and its interaction with Hsp90 exerts an anti-tumor effect on human melanomas by modulating the C-Raf activity, especially when combined with Hsp90 inhibitors. These findings may provide hints for therapeutic approaches that could be beneficial for improving the treatment of human melanoma.

Supplementary Materials

The following supporting information can be downloaded at: https://www.mdpi.com/article/10.3390/biomedicines10102551/s1.

Author Contributions

E.G., F.P. and F.T. designed the experiments; E.G. and F.P. performed research experiments; E.G., F.P. and F.T. wrote the paper. All authors critically reviewed the manuscript and approved the final version. All authors have read and agreed to the published version of the manuscript.

Funding

This study was partly supported by a generous donation from the Stillitani family in the loving memory of Carmelo.

Institutional Review Board Statement

Animal experiments were conducted under the Ohio State University guidelines established for the Animal Facility. The protocol was approved by the Institutional Animal Care and Use Committee (IACUC) at Ohio State University (IACUC protocol number: 2010A00000146; approval date: 15 August 2011).

Informed Consent Statement

Not applicable.

Data Availability Statement

The data can be requested by emailing the corresponding authors.

Acknowledgments

This work is dedicated to the memory of Pietro Gaudio, who died on 11 March 1996. We are very grateful to Carlo M. Croce for his logistical support, Zhenghua Luo for his technical support, and Kari Green-Church for mass spectrometry analysis support. The authors are also indebted to Luisa Infante for her precious support in editing this manuscript.

Conflicts of Interest

The authors declare no conflict of interest.

References

  1. Ohta, M.; Inoue, H.; Cotticelli, M.G.; Kastury, K.; Baffa, R.; Palazzo, J.; Siprashvili, Z.; Mori, M.; McCue, P.; Druck, T.; et al. The FHIT gene, spanning the chromosome 3p14.2 fragile site and renal carcinoma-associated t(3;8) breakpoint, is abnormal in digestive tract cancers. Cell 1996, 84, 587–597. [Google Scholar] [CrossRef] [Green Version]
  2. Trapasso, F.; Krakowiak, A.; Cesari, R.; Arkles, J.; Yendamuri, S.; Ishii, H.; Vecchione, A.; Kuroki, T.; Bieganowski, P.; Pace, H.C.; et al. Designed FHIT alleles establish that Fhit-induced apoptosis in cancer cells is limited by substrate binding. Proc. Natl. Acad. Sci. USA 2003, 100, 1592–1597. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  3. Dumon, K.R.; Ishii, H.; Fong, L.Y.; Zanesi, N.; Fidanza, V.; Mancini, R.; Vecchione, A.; Baffa, R.; Trapasso, F.; During, M.J.; et al. FHIT gene therapy prevents tumor development in Fhit-deficient mice. Proc. Natl. Acad. Sci. USA 2001, 98, 3346–3351. [Google Scholar] [CrossRef] [Green Version]
  4. Druck, T.; Cheung, D.G.; Park, D.; Trapasso, F.; Pichiorri, F.; Gaspari, M.; Palumbo, T.; Aqeilan, R.I.; Gaudio, E.; Okumura, H.; et al. Fhit-Fdxr interaction in the mitochondria: Modulation of reactive oxygen species generation and apoptosis in cancer cells. Cell Death Dis. 2019, 10, 147. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  5. Gaudio, E.; Paduano, F.; Spizzo, R.; Ngankeu, A.; Zanesi, N.; Gaspari, M.; Ortuso, F.; Lovat, F.; Rock, J.; Hill, G.A.; et al. Fhit delocalizes annexin a4 from plasma membrane to cytosol and sensitizes lung cancer cells to paclitaxel. PLoS ONE 2013, 8, e78610. [Google Scholar] [CrossRef] [PubMed]
  6. Gaudio, E.; Paduano, F.; Ngankeu, A.; Ortuso, F.; Lovat, F.; Pinton, S.; D’Agostino, S.; Zanesi, N.; Aqeilan, R.I.; Campiglia, P.; et al. A Fhit-mimetic peptide suppresses annexin A4-mediated chemoresistance to paclitaxel in lung cancer cells. Oncotarget 2016, 7, 29927–29936. [Google Scholar] [CrossRef] [Green Version]
  7. Gaudio, E.; Paduano, F.; Croce, C.M.; Trapasso, F. The Fhit protein: An opportunity to overcome chemoresistance. Aging 2016, 8, 3147–3150. [Google Scholar] [CrossRef] [Green Version]
  8. Moulin, A.P.; Clément, G.; Bosman, F.T.; Zografos, L.; Benhattar, J. Methylation of CpG island promoters in uveal melanoma. Br. J. Ophthalmol. 2008, 92, 281–285. [Google Scholar] [CrossRef] [Green Version]
  9. Scholes, A.G.; Liloglou, T.; Maloney, P.; Hagan, S.; Nunn, J.; Hiscott, P.; Damato, B.E.; Grierson, I.; Field, J.K. Loss of heterozygosity on chromosomes 3, 9, 13, and 17, including the retinoblastoma locus, in uveal melanoma. Investig. Ophthalmol. Vis. Sci. 2001, 42, 2472–2477. [Google Scholar]
  10. Wu, S.; Singh, R.K. Resistance to chemotherapy and molecularly targeted therapies: Rationale for combination therapy in malignant melanoma. Curr. Mol. Med. 2011, 11, 553–563. [Google Scholar] [CrossRef] [Green Version]
  11. Paluncic, J.; Kovacevic, Z.; Jansson, P.J.; Kalinowski, D.; Merlot, A.M.; Huang, M.L.-H.; Lok, H.C.; Sahni, S.; Lane, D.J.; Richardson, D.R. Roads to melanoma: Key pathways and emerging players in melanoma progression and oncogenic signaling. Biochim. Biophys. Acta-Mol. Cell Res. 2016, 1863, 770–784. [Google Scholar] [CrossRef] [PubMed]
  12. Leonardi, G.C.; Falzone, L.; Salemi, R.; Zanghì, A.; Spandidos, D.A.; Mccubrey, J.A.; Candido, S.; Libra, M. Cutaneous melanoma: From pathogenesis to therapy. Int. J. Oncol. 2018, 52, 1071–1080. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  13. Jenkins, R.W.; Fisher, D.E. Treatment of advanced melanoma in 2020 and beyond. J. Investig. Dermatol. 2021, 141, 23–31. [Google Scholar] [CrossRef] [PubMed]
  14. Palmieri, G.; Ombra, M.; Colombino, M.; Casula, M.; Sini, M.; Manca, A.; Paliogiannis, P.; Ascierto, P.A.; Cossu, A. Multiple molecular pathways in melanomagenesis: Characterization of therapeutic targets. Front. Oncol. 2015, 5, 183. [Google Scholar] [CrossRef]
  15. Czarnecka, A.M.; Bartnik, E.; Fiedorowicz, M.; Rutkowski, P. Targeted therapy in melanoma and mechanisms of resistance. Int. J. Mol. Sci. 2020, 21, 4576. [Google Scholar] [CrossRef]
  16. Arslanbaeva, L.R.; Santoro, M.M. Adaptive redox homeostasis in cutaneous melanoma. Redox Biol. 2020, 37, 101753. [Google Scholar] [CrossRef]
  17. Ortuso, F.; Paduano, F.; Carotenuto, A.; Gomez-Monterrey, I.; Bilotta, A.; Gaudio, E.; Sala, M.; Artese, A.; Vernieri, E.; Dattilo, V.; et al. Discovery of PTPRJ agonist peptides that effectively inhibit in vitro cancer cell proliferation and tube formation. ACS Chem. Biol. 2013, 8, 1497–1506. [Google Scholar] [CrossRef]
  18. Paduano, F.; Dattilo, V.; Narciso, D.; Bilotta, A.; Gaudio, E.; Menniti, M.; Agosti, V.; Palmieri, C.; Perrotti, N.; Fusco, A.; et al. Protein tyrosine phosphatase PTPRJ is negatively regulated by microRNA-328. FEBS J. 2013, 280, 401–412. [Google Scholar] [CrossRef]
  19. Paduano, F.; Ortuso, F.; Campiglia, P.; Raso, C.; Iaccino, E.; Gaspari, M.; Gaudio, E.; Mangone, G.; Carotenuto, A.; Bilotta, A.; et al. Isolation and functional characterization of peptide agonists of PTPRJ, a tyrosine phosphatase receptor endowed with tumor suppressor activity. ACS Chem. Biol. 2012, 7, 1666–1676. [Google Scholar] [CrossRef]
  20. Gaudio, E.; Paduano, F.; Ngankeu, A.; Lovat, F.; Fabbri, M.; Sun, H.-L.; Gasparini, P.; Efanov, A.; Peng, Y.; Zanesi, N. Heat shock protein 70 regulates Tcl1 expression in leukemia and lymphomas. Blood J. Am. Soc. Hematol. 2013, 121, 351–359. [Google Scholar] [CrossRef] [Green Version]
  21. Gaudio, E.; Spizzo, R.; Paduano, F.; Luo, Z.; Efanov, A.; Palamarchuk, A.; Leber, A.S.; Kaou, M.; Zanesi, N.; Bottoni, A. Tcl1 interacts with Atm and enhances NF-κB activation in hematologic malignancies. Blood J. Am. Soc. Hematol. 2012, 119, 180–187. [Google Scholar] [CrossRef] [PubMed]
  22. Trapasso, F.; Yendamuri, S.; Dumon, K.R.; Iuliano, R.; Cesari, R.; Feig, B.; Seto, R.; Infante, L.; Ishii, H.; Vecchione, A. Restoration of receptor-type protein tyrosine phosphatase η function inhibits human pancreatic carcinoma cell growth in vitro and in vivo. Carcinogenesis 2004, 25, 2107–2114. [Google Scholar] [CrossRef]
  23. Maloney, A.; Workman, P. HSP90 as a new therapeutic target for cancer therapy: The story unfolds. Expert Opin. Biol. Ther. 2002, 2, 3–24. [Google Scholar] [CrossRef]
  24. Neckers, L. Chaperoning oncogenes: Hsp90 as a target of geldanamycin. Handb. Exp. Pharmacol. 2006, 172, 259–277. [Google Scholar] [CrossRef]
  25. Chiosis, G. Targeting chaperones in transformed systems--a focus on Hsp90 and cancer. Expert Opin. Ther. Targets 2006, 10, 37–50. [Google Scholar] [CrossRef]
  26. Whitesell, L.; Lindquist, S.L. HSP90 and the chaperoning of cancer. Nat. Rev. Cancer 2005, 5, 761–772. [Google Scholar] [CrossRef] [PubMed]
  27. Schulte, T.W.; An, W.G.; Neckers, L.M. Geldanamycin-induced destabilization of Raf-1 involves the proteasome. Biochem. Biophys. Res. Commun. 1997, 239, 655–659. [Google Scholar] [CrossRef] [PubMed]
  28. Isaacs, J.S.; Xu, W.; Neckers, L. Heat shock protein 90 as a molecular target for cancer therapeutics. Cancer Cell 2003, 3, 213–217. [Google Scholar] [CrossRef] [Green Version]
  29. Wellbrock, C.; Karasarides, M.; Marais, R. The RAF proteins take centre stage. Nat. Rev. Mol. Cell Biol. 2004, 5, 875–885. [Google Scholar] [CrossRef] [PubMed]
  30. Schulte, T.W.; Blagosklonny, M.V.; Romanova, L.; Mushinski, J.F.; Monia, B.P.; Johnston, J.F.; Nguyen, P.; Trepel, J.; Neckers, L.M. Destabilization of Raf-1 by geldanamycin leads to disruption of the Raf-1-MEK-mitogen-activated protein kinase signalling pathway. Mol. Cell. Biol. 1996, 16, 5839–5845. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  31. Carr, S.; Smith, C.; Wernberg, J. Epidemiology and Risk Factors of Melanoma. Surg. Clin. North Am. 2020, 100, 1–12. [Google Scholar] [CrossRef]
  32. Trapasso, F.; Pichiorri, F.; Gaspari, M.; Palumbo, T.; Aqeilan, R.I.; Gaudio, E.; Okumura, H.; Iuliano, R.; Di Leva, G.; Fabbri, M.; et al. Fhit interaction with ferredoxin reductase triggers generation of reactive oxygen species and apoptosis of cancer cells. J. Biol. Chem. 2017, 292, 14279. [Google Scholar] [CrossRef]
Figure 1. Hsp90 interacts with Fhit. (A) Proteins derived from Colo38 were immunoprecipitated using an agarose-conjugated antibody against IgG or Hsp90, and immunoprecipitates were separated on SDS-PAGE and subsequently blotted with Fhit and Hsp90 antibodies. (B) Fhit protein expression was evaluated with normal melanocyte and melanoma cancer cells via W.B. with an antibody against Fhit. Gapdh and B-actin were used as loading controls.
Figure 1. Hsp90 interacts with Fhit. (A) Proteins derived from Colo38 were immunoprecipitated using an agarose-conjugated antibody against IgG or Hsp90, and immunoprecipitates were separated on SDS-PAGE and subsequently blotted with Fhit and Hsp90 antibodies. (B) Fhit protein expression was evaluated with normal melanocyte and melanoma cancer cells via W.B. with an antibody against Fhit. Gapdh and B-actin were used as loading controls.
Biomedicines 10 02551 g001
Figure 2. Fhit affects C-Raf expression through Hsp90. (A,B) Colo38 and CHL-1 melanoma cell lines were transfected with an empty vector or Fhit and treated with 17-AAG or DMSO at the indicated concentration for 24 hrs. Then, cell lysate was separated on SDS-PAGE gel and blotted with B-Raf, C-Raf, p-ERK, ERK, Fhit, and GAPDH antibodies.
Figure 2. Fhit affects C-Raf expression through Hsp90. (A,B) Colo38 and CHL-1 melanoma cell lines were transfected with an empty vector or Fhit and treated with 17-AAG or DMSO at the indicated concentration for 24 hrs. Then, cell lysate was separated on SDS-PAGE gel and blotted with B-Raf, C-Raf, p-ERK, ERK, Fhit, and GAPDH antibodies.
Biomedicines 10 02551 g002
Figure 3. Restoration of Fhit expression reduces cell proliferation and induces apoptosis in human melanoma. (A) E6LCP, Colo38 and CHL1, AdGFP, AdFHIT, or mock-infected cells were analyzed using MTS assays (Student’s t-test, * p < 0.05; n.s., not significant). (B) Colo38 AdFHIT and AdGFP, untreated or treated with 17-AAG at 2 μM, were analyzed using a TUNEL assay (one-way ANOVA, *** p < 0.001). (C) Cell lysates from the experiment described in B were blotted with antibodies against Fhit, Parp, and Gapdh. (D) Colo38 AdGFP, AdFHIT, or mock-infected cells, untreated or treated with 17-AAG at 2 μM, were counted, and cell growth curves were calculated. Results show means ± SD from 3 independent experiments (Student’s t-test, * p < 0.05).
Figure 3. Restoration of Fhit expression reduces cell proliferation and induces apoptosis in human melanoma. (A) E6LCP, Colo38 and CHL1, AdGFP, AdFHIT, or mock-infected cells were analyzed using MTS assays (Student’s t-test, * p < 0.05; n.s., not significant). (B) Colo38 AdFHIT and AdGFP, untreated or treated with 17-AAG at 2 μM, were analyzed using a TUNEL assay (one-way ANOVA, *** p < 0.001). (C) Cell lysates from the experiment described in B were blotted with antibodies against Fhit, Parp, and Gapdh. (D) Colo38 AdGFP, AdFHIT, or mock-infected cells, untreated or treated with 17-AAG at 2 μM, were counted, and cell growth curves were calculated. Results show means ± SD from 3 independent experiments (Student’s t-test, * p < 0.05).
Biomedicines 10 02551 g003
Figure 4. Fhit regulates C-Raf expression in melanoma cells. (A) C-Raf, Ki67, and H&E were analyzed through immunohistochemistry in mock- and FHIT-transfected Colo38 melanoma cells. (B) Colo38 melanoma cells, mock- or FHIT-transfected, were treated with DMSO, Sorafenib, and PLX4032. Then, cell lysates were immunoblotted with C-Raf, p-C-RAF, FHIT, and GAPDH antibodies.
Figure 4. Fhit regulates C-Raf expression in melanoma cells. (A) C-Raf, Ki67, and H&E were analyzed through immunohistochemistry in mock- and FHIT-transfected Colo38 melanoma cells. (B) Colo38 melanoma cells, mock- or FHIT-transfected, were treated with DMSO, Sorafenib, and PLX4032. Then, cell lysates were immunoblotted with C-Raf, p-C-RAF, FHIT, and GAPDH antibodies.
Biomedicines 10 02551 g004
Figure 5. Tumor suppressor activity of Fhit in melanoma xenograft. (A) Tumor volumes at 10, 20, 30, and 40 days for the mock, AdGFP, and AdFHIT groups of mice (5 mice/group). Overall, 10 × 106 Colo38 cells were pre-infected with AdFHIT or AdGFP for 12 h and then injected subcutaneously into nude mice (5 mice for the group). Tumors were measured every 10 days, as reported in the graph (Student’s t-test, * p < 0.05). (B) Tumors were extracted and weighed after 40 days (Student’s t-test, * p < 0.05).
Figure 5. Tumor suppressor activity of Fhit in melanoma xenograft. (A) Tumor volumes at 10, 20, 30, and 40 days for the mock, AdGFP, and AdFHIT groups of mice (5 mice/group). Overall, 10 × 106 Colo38 cells were pre-infected with AdFHIT or AdGFP for 12 h and then injected subcutaneously into nude mice (5 mice for the group). Tumors were measured every 10 days, as reported in the graph (Student’s t-test, * p < 0.05). (B) Tumors were extracted and weighed after 40 days (Student’s t-test, * p < 0.05).
Biomedicines 10 02551 g005
Publisher’s Note: MDPI stays neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Share and Cite

MDPI and ACS Style

Paduano, F.; Gaudio, E.; Trapasso, F. The Tumour Suppressor Fhit Protein Activates C-Raf Ubiquitination and Degradation in Human Melanoma Cells by Interacting with Hsp90. Biomedicines 2022, 10, 2551. https://doi.org/10.3390/biomedicines10102551

AMA Style

Paduano F, Gaudio E, Trapasso F. The Tumour Suppressor Fhit Protein Activates C-Raf Ubiquitination and Degradation in Human Melanoma Cells by Interacting with Hsp90. Biomedicines. 2022; 10(10):2551. https://doi.org/10.3390/biomedicines10102551

Chicago/Turabian Style

Paduano, Francesco, Eugenio Gaudio, and Francesco Trapasso. 2022. "The Tumour Suppressor Fhit Protein Activates C-Raf Ubiquitination and Degradation in Human Melanoma Cells by Interacting with Hsp90" Biomedicines 10, no. 10: 2551. https://doi.org/10.3390/biomedicines10102551

Note that from the first issue of 2016, this journal uses article numbers instead of page numbers. See further details here.

Article Metrics

Back to TopTop