Reduced expression of secretogranin VGF in laryngeal squamous cell carcinoma

  • Authors:
    • Francesca Gabanella
    • Daniela Maftei
    • Andrea Colizza
    • Emma Rullo
    • Mara Riminucci
    • Elena Pasqualucci
    • Maria Grazia Di Certo
    • Roberta Lattanzi
    • Roberta Possenti
    • Alessandro Corsi
    • Antonio Greco
    • Marco De Vincentiis
    • Cinzia Severini
    • Massimo Ralli
  • View Affiliations

  • Published online on: November 30, 2023     https://doi.org/10.3892/ol.2023.14170
  • Article Number: 37
Metrics: Total Views: 0 (Spandidos Publications: | PMC Statistics: )
Total PDF Downloads: 0 (Spandidos Publications: | PMC Statistics: )


Abstract

Laryngeal cancer accounts for one‑third of all head and neck tumors, with squamous cell carcinoma (SCC) being the most predominant type, followed by neuroendocrine tumors. Chromogranins, are commonly used as biomarkers for neuroendocrine tumors, including laryngeal cancer. It has been reported that secretogranin VGF, a member of the chromogranin family, can be also used as a significant biomarker for neuroendocrine tumors. However, the expression and role of VGF in laryngeal carcinomas have not been previously investigated. Therefore, the present study aimed to determine the expression levels of VGF in laryngeal SCC (LSCC). The present study collected tumor tissues, as well as serum samples, from a cohort of 15 patients with LSCC. The results of reverse transcription‑quantitative PCR, western blot analysis and immunofluorescence assays showed that the selective VGF precursor was downregulated in patients with LSCC. Notably, in tumor tissue, the immunoreactivity for VGF was found in vimentin‑positive cells, probably corresponding to T lymphocytes. The current preliminary study suggested that the reduced expression levels of VGF observed in tumor tissue and at the systemic level could sustain LSCC phenotype. Overall, VGF could be a potential biomarker for detecting neoplastic lesions with a higher risk of tumor invasiveness, even in non‑neuroendocrine tumors.

Introduction

Laryngeal cancer, characterized by an increasing annual incidence, is the second most common head and neck type of cancer, accounting for ~20% of all head and neck cancer cases (1,2). According to the 2018 Global Cancer Statistics report, the incidence of laryngeal cancer was 2/100,000 individuals, with a mortality rate of 1/100,000 (3).

Laryngeal cancers are predominantly (95%) squamous cell carcinomas (SCC), followed by neuroendocrine neoplasms of the larynx, which are the most common non-squamous tumors of the larynx, despite their rarity (4,5).

Comprehensive treatment approaches, such as surgery, radiotherapy, chemotherapy and concurrent chemotherapy and radiotherapy have provided a higher 5-year survival rate (50–80%) in patients with laryngeal cancer (6). However, despite the current therapeutic advances, the survival rate of patients with laryngeal cancer remains poor due to the advanced stage of diagnosis, the high tumor recurrence rate, and distant metastases (7).

Early diagnosis serves a crucial role in the early detection of relapses and in reducing mortality via enhancing the effectiveness of the currently available therapeutic approaches. Therefore, identifying effective diagnostic and prognostic biomarkers for laryngeal cancer is critical to guide disease management and improve treatment outcomes.

The members of the chromogranin family, and more particular chromogranin A (CGA) and its proteolytically derived peptides, are widely used to identify particular types of tumors with a neuroendocrine-like phenotype (8) and allow the assessment of the malignancy grade and metastatic potential of tumors. Bartolomucci et al (9) demonstrated that CGA is expressed in several types of endocrine and neuroendocrine tumors, such as prostate cancer (10), gastrointestinal neuroendocrine tumors (11) and neuroendocrine carcinomas of the head and neck, including laryngeal cancer (1113).

In addition to CGA, other secretogranins have been also identified as potential endocrine tumor markers (14), including the VGF (non-acronymic) polypeptide, identified from the ‘V’ clone of the PC12 cDNA library (15).

In humans, VGF encodes a precursor protein (pro-VGF), which produces several peptides involved in food intake, energy balance and metabolism, water and electrolyte homeostasis, reproduction, pain, learning and memory (16). In addition, it has been reported that pro-VGF can promote neuronal growth and prevent apoptosis (17,18), while it serves a significant role in the pathogenesis of several types of neuroendocrine tumors (1921).

In addition to neuroendocrine tumors, previous studies suggest that VGF could exhibit anticancer effects in non-endocrine tumors, such as breast (22), testicular (23) and ovarian cancer (24), thus indicating that VGF could be a potential biomarker in the above types of cancer. Therefore, dysregulation of VGF expression and processing could be dependent on tumor type.

As studies regarding the expression and role of VGF in laryngeal cancer are lacking, the present study aimed to investigate the expression profile of VGF in LSCC tumor tissues. In addition, since a previous study indicated that CGA levels in the blood of patients with endocrine tumors could act as a potential biomarker (25), the measurement of VGF in blood could provide an additional tool for the diagnosis and monitoring of laryngeal tumors.

Materials and methods

Patients

A total of 15 patients with LSCC were included in the present study. The protocol conformed to the Declaration of Helsinki and its later amendments and was approved by the internal Institutional Review Board (Ethical Committee of Sapienza University and Policlinico Umberto I, Rome, Italy; approval number: 6129).

The sites of the tumors and staging and grading were established according to the American Joint Committee on Cancer (26). Archival formalin-fixed paraffin-embedded (FFPE) tumor tissue from a laryngeal neuroendocrine carcinoma was used as positive control in the immunofluorescence experiments. Serum from five age-matched healthy subjects was used to quantitate VGF by western blotting for comparison with that of five of the patients with LSCC included in the present study. A clinical synopsis of the patients with LSCC included in the present study is in Table I.

Table I.

Clinical synopsis of the patients with laryngeal squamous cell carcinoma included in the present study.

Table I.

Clinical synopsis of the patients with laryngeal squamous cell carcinoma included in the present study.

Patient numberSexAgeTumor locationpTNM stageAmerican Joint Committee on Cancer stageGrade
1Male79GlottispT4aN0M0IVaG2
2Male78SupraglottispT3N3bM0IVbG2
3Male77SupraglottispT4aN0M0IVaG2
4Male61SupraglottispT4aN1M0IVaG3
5Female74GlottispT4aN0M0IVaG2
6Male63GlottispT4aN2aM0IVaG2
7Male71GlottispT3N0M0IIIG2
8Male56SupraglottispT4aN1M0IVaG2
9Male58GlottispT3N3bM0IVbG2
10Female75SupraglottispT3N0M0IIIG2
11Male73SupraglottispT3N3bM0IVbG2
12Female77GlottispT3N0M0IIIG2
13Male63GlottispT4aN0M0IVaG2
14Male78SupraglottispT3N3bM0IVbG2
15Male78GlottispT3N0M0IIIG2
Reagents

The following antibodies were used: Anti-VGF mouse monoclonal antibody (cat. no. sc-515482; Santa Cruz Biotechnology, Inc.); dilution for immunofluorescence, 1:100; dilution for western blotting (WB), 1:500; anti-GAPDH mouse monoclonal antibody (cat. no. sc-47724; Santa Cruz Biotechnology, Inc.; dilution for WB, 1:500); anti-Vimentin rabbit monoclonal antibody (cat. no. ab92547; Abcam; dilution for immunofluorescence, 1:500); anti-CD3 monoclonal antibody (cat. no. ab699; Abcam; dilution for immunofluorescence, 1:100). The secondary antibodies conjugated to horseradish peroxidase were purchased from Jackson ImmunoResearch Laboratories (cat. no. 111-035-003; cat. no. 115-035-003) and used at a dilution of 1:5,000. The Alexa Fluor488- and Alexa Fluor594-conjugated secondary antibodies were purchased from Thermo Fisher Scientific Inc. (cat. nos. A-11029 and A-11012) and were used at a dilution of 1:250. TRIzol® was purchased from Thermo Fisher Scientific Inc. Complete protease and phosphatase inhibitor cocktail (cOmplete, EDTA-free Protease and PhosSTOP tablets) were from Roche Diagnostics and the Chemiluminescence ECL kit was from Cytiva. Ponceau S Staining Solution and ProLong with DAPI were from Thermo Fisher Scientific Inc.

RNA extraction, retro-transcription and reverse transcription-quantitative (RT-q) PCR

The total RNA from frozen tumor and adjacent non-tumor tissue samples were extracted using TRIzol® reagent according to the manufacturer's instructions and was then reverse transcribed using a High-Capacity cDNA Reverse Transcription kit (Thermo Fisher Scientific). qPCR was performed using an iCycler Detection System (Bio-Rad Laboratories, Inc.). The cDNAs were amplified using iQ SYBR Green Supermix (Bioline; Meridian Bioscience) and specific sense and antisense human primers for the interest gene: VGF (Eurofins Genomics). Each reaction was performed in triplicate under the same thermal cycling conditions as follows: 95°C for 10 min, followed by 40 cycles at 95°C for 30 sec, 60°C and 72°C for 30 sec, to obtain the cycle time (Ct) mean. A reaction mixture without cDNA was used as control and post-amplification dissociation curves were performed to verify the presence of a single amplification product and the absence of genomic DNA contamination. The Ct mean value of the target gene was normalized to the Ct mean value of the house-keeping gene, 18S rRNA, and the comparative method (2-∆∆Cq) (27) was obtained for each patient using gene expression value of normal tissues as calibrator. Data were reported as fold increase of the target gene mRNA compared to the normal tissues. The human primer sequences used in the present study were: VGF: F: 5′-AGCATAAAGAGCCGGTAGCC-3′, R: 5′-GGAAAAGCTCTCCCTCGTCC-3′; 18SrRNA F: 5′-ACCGGGTTGGTTTTGATCTG-3′, R: 5′-ATCCTGCCAGTAGCATATGC-3′.

Protein extraction

Protein extraction was carried out as previously described (28). Briefly, frozen tumor and adjacent non-tumor tissue samples were processed in lysis buffer (1% SDS; 1% NP-40, 5% glycerol and 5 mM EDTA) supplemented with complete protease and phosphatase inhibitor cocktail using a homogenizer (7 mm, OMNI International). Samples were boiled for 10 min and centrifuged for 20 min at 12,000 × g at 4°C. Supernatants were collected and protein concentration was measured by a Qubit fluorometer (Invitrogen; Thermo Fisher Scientific, Inc.), according to the manufacturer's instructions. Protein extracts were stored at −80°C until use.

WB analysis

WB analysis was carried out as previously described (28). Briefly, protein extracts (30 µg/lane) were electrophoresed through 10% SDS-PAGE and transferred onto nitrocellulose membranes (Cytiva). The membrane was stained with Ponceau Solution for 5 min at room temperature, and then washed. After blocking the proteins with 4% non-fat died milk (PanReac; AppliChem) for 2 h at room temperature, the primary antibodies incubation was performed overnight at 4°C. Membranes were then washed with PBS three times for 10 min and incubated with the secondary HRP-conjugated antibodies for 40 min at room temperature. After three washes in PBS, immunodetection of the reactive bands was revealed by chemiluminescence (ECL kit; Cytiva) and analyzed by iBright 1500 (Thermo Fisher Scientific Inc.). ImageJ v1.53a (National Institutes of Health) was used for densiometric analysis.

Immunofluorescence

Immunofluorescence analysis of FFPE samples was performed as described previously (28). Briefly, paraffin-embedded sections were dewaxed by two changes of xylene (5 min each) and hydrated in graded ethanol solutions (100, 90, and 70%, ethanol, for 2 min each). Sections were incubated in the antigen retrieval solution (10 mM sodium citrate, 0.05% Tween 20, pH 6.1) for 3×2 min and 4×30 sec into a microwave oven at 750 W. After cooling to room temperature slides were rinsed in PBS and blocked with 1% BSA in PBS for 1 h at room temperature. Samples were incubated at 4°C overnight using the appropriate primary antibodies; washed three times in PBS/0.1% Tween 20; and incubated at room temperature with the appropriate secondary antibodies for 1 h. Slides were mounted with ProLong with DAPI (Thermo Fisher Scientific, Inc.) and examined by an epifluorescence microscope (Olympus BX53; Olympus Corporation) equipped with a SPOT RT3 camera. Images were merged using the image analysis software IAS 2000 (Delta Sistemi).

Blood collection

Peripheral blood samples of 5 ml were available from 5 of 15 LSCC patients included in the present study, and 5 age-matched healthy subjects. The samples were collected in BD Vacutainer Serum Separation Tubes (BD Biosciences) and centrifuged at 1,000 × g for 15 min at 4°C to separate serum from plasma. Serum was then stored at −80°C, until use.

Statistical analysis

All experiments were performed for at least three independent replicates. Data are presented as mean ± standard deviation Statistical analysis was performed using GraphPad Prism 9.4.1 software (GraphPad Software; Dotmatics). Data were analyzed using both the unpaired and paired t-test. P<0.01 was considered to indicate a statistically significant difference.

Results

Patients

A total of 15 patients with LSCC were enrolled in the present study. Among them, 11 patients provided tissue samples for WB and qPCR analysis, four patients for immunofluorescence staining and five patients for serum analysis. The clinical characteristics of patients with LSCC are listed in Table I.

Expression of VGF in LSCC tissues

To evaluate the expression profile of VGF in LSCC tissues, its mRNA and protein expression levels were detected by qPCR and WB, respectively (n=11 subjects for each assay). For each patient, the expression levels of VGF in LSCC tissues (T) were compared with those in normal adjacent tissues (CTR). As shown in Fig. 1, no significant differences were observed in the amount of VGF mRNA in tumor samples compared with CTR. In Fig. 2A, representative immunoblots obtained from 11 patients with LSCC are shown. WB revealed the presence of an immunoreactive band with a molecular weight of ~70 kDa, corresponding to human pro-VGF (19). Pro-VGF was mainly detected in CTR. WB was carried out using samples derived from 11 patients with LSCC and the expression levels of VGF were normalized to those of GAPDH. The results showed that pro-VGF was downregulated in tumor samples compared with CTR (Fig. 2B; P<0.01). In the current study, the expression levels of other pro-VGF-related peptides were not detected.

Localization of VGF in laryngeal tumor tissues

Immunofluorescence staining of tissues from primitive LSCC and LSCC with lymph node metastases was performed to evaluate the localization of VGF in SCC. Sections from FFPE laryngeal neuroendocrine carcinoma tissue samples served as a positive control. The neoplastic cells, as expected, were negative for vimentin. The cells immunoreactive for vimentin are ‘stromal cells’ (i.e., they are distributed among the nests) (29). As expected, a strong VGF immunoreactivity was observed in laryngeal neuroendocrine carcinoma tissues (Fig. 3A). By contrast, no immunoreactivity was obtained in LSCC tissues (Fig. 3B) and LSCC tissues with lymph node metastasis (Fig. 3C). However, a moderate immunoreactivity was detected around and within the SCC nests, co-localizing with vimentin, possibly representing tumor-(Fig. 3B) and nodal-related (Fig. 3C) T-lymphocytes (30,31). SCC tissues were also subjected to dual immunostaining for CD3 (T-lymphocytes marker) and vimentin (Fig. 3D). As expected, a CD3 immunostaining (green) was observed around and within the tumoral nests, co-localizing with vimentin (red; Fig. 3D), confirming the presence of T-lymphocytes. No immunoreactivity for VGF was observed in the epithelial lining of the larynx in sections from both LSCC and positive control tissues. However, unavailability of images of adjacent non-tumor tissue staining represents a limitation of the present study.

VGF levels in the serum of patients with LSCC

Since the protein expression levels of pro-VGF were decreased in tissues derived from patients with LSCC compared with CTR, the VGF content in the serum derived from a subgroup of five patients with LSCC were detected by WB. The serum levels of VGF in patients with LSCC were compared with those in age-matched healthy subjects. As shown in Fig. 4B, the levels of VGF-related peptide (~70 kDa) were significantly reduced in the serum from patients with LSCC compared with those in the serum of healthy donors (Fig. 4C; P<0.01).

Discussion

The present pilot study aimed to investigate the expression and the putative role, if any, of VGF in LSCC. Therefore, the expression profile of VGF-derived peptides in tumor tissues and serum of patients with LSCC was determined.

As expected, the results indicated that, at least in the larynx, SCC cells did not express VGF. This finding was consistent with that obtained in a previous study showing that only a very small fraction of head and neck SCCs could express neuroendocrine markers (12).

VGF is an active neuroendocrine regulatory polypeptide, mainly expressed in the human hypothalamus, in the medial and lateral frontal gyrus and in several neuroendocrine tissues, including the pituitary gland and various gastrointestinal and pancreatic neuroendocrine cells (19). In addition to nerve growth factor, several stimuli can induce VGF expression, such as cell depolarization, growth factors, IL-6, insulin and cyclic adenosine monophosphate (19).

The VGF gene encodes a precursor protein, namely pro-VGF, with a molecular weight of ~70 kDa, which in humans consists of 615 amino acids (19). Pro-VGF is then processed by pro-protein convertases (PC1/3 or PC2), resulting in a series of VGF-related peptide fragments, which are stored in dense core granules and secreted via regulated pathways (32). It has been reported that several low molecular weight VGF-encoded peptides, covering ~20% of the pro-VGF sequence, including TLQP-21, TLQP-62 and AQEE-30, with total lengths of 21, 62 and 30 amino acids, respectively, exhibit several biological functions (15,33,34). Processing at different sites or under diverse conditions can result in different acting end products. However, the significance of VGF remains currently poorly understood (19).

Rindi et al (35) demonstrated that the expression of VGF-related peptides, such as that of pro-VGF, in human neuroendocrine cells could promote endocrine hyperplasia and neoplasia, depending on the cell type-specific processing of pro-VGF. This finding was further supported by the finding that 88/102 endocrine tumors tested were positive for the expression of VGF peptides, thus indicating that VGF could mark an active/proliferative state in response to specific stimuli (35). The increased expression and release of VGF-related fragment peptides have been also verified in large-cell neuroendocrine carcinoma of the lungs (21,36) and in breast cancer with neuroendocrine features (20). The present study also demonstrated that, in addition to CGA, VGF was also upregulated in neuroendocrine carcinomas of the larynx, thus suggesting that VGF could be considered as a potential novel biomarker for neuroendocrine tumors.

However, emerging evidence has also suggested that VGF exhibits different roles, as it possesses a protective effect on non-endocrine tumors, such as breast (22), testicular (23) and ovarian cancer (24). Therefore, it was hypothesized that the abnormal expression and processing of VGF could depend on tumor type.

RT-qPCR analysis revealed that the mRNA expression levels of VGF were comparable in LSCC tissues compared with the adjacent non-tumor tissues. By contrast, WB showed that the protein expression levels of pro-VGF were significantly reduced in LSCC tissues compared with CTR tissues. No other lower molecular weight bands were present in the membrane, at least not under the experimental conditions of the present study.

In addition, immunofluorescence assays verified the weak VGF immunoreactivity in primary LSCC and LSCC with lymph node metastasis. Notably, VGF immunoreactivity was observed in vimentin-positive cells within the stromal tissue in tumor samples, possibly corresponding to T-lymphocytes (30,31). Pro-VGF levels were also notably reduced in the serum of patients with LSCC compared with healthy donors, thus indicating that VGF could be downregulated both locally and systemically through post-transcriptional mechanisms.

The present results, obtained in the serum of a limited number of LSCC patients using WB, should be confirmed by quantifying the level of VGF in a larger number of patients, possibly using a more selective analysis, such as an ELISA test.

The current study also aimed to uncover the meaning of VGF downregulation in tissues and serum derived from patients with LSCC, as the significance of VGF precursor in the diagnosis or prognosis of patients is worth further exploration. Indeed, additional in vivo and in vitro experiments could confirm these preliminary findings and support evidence of abnormal expression and processing of VGF in this type of tumor. By using cellular models of human laryngeal carcinoma, it is hoped to provide further evidence of the VGF implication in cancer-relevant behaviors.

Given the role of VGF in regulating energy homeostasis and metabolism, it was hypothesized that VGF depletion in various types of tumor, such as LSCC, could promote the proliferation and spread of neoplastic cells. Indeed, previous studies show that VGF knockout mice are hyperactive and hypermetabolic (37). Hypermetabolism is a well-known feature of cancer, which allows tumor cells to undergo uncontrolled cell division and proliferation (38).

Growing evidence has also supported the significance of enhanced metabolism and thus energy production in the tumor microenvironment. The above effect negatively affects the availability of nutrients to immune cells, and more particular in tumor-invasive T cells, which also require a high metabolic energy status to function efficiently. Therefore, immune cells should compete with cancer cells for the available energy resources (39). However, whether VGF downregulation in SCC also serves a significant role in other sites either within or outside the head and neck region should be further investigated.

Acknowledgements

Not applicable.

Funding

The present study was supported by Medio Progetto di Ateneo 2019 (grant no. RM11916B7E5A0D4) to Massimo Ralli.

Availability of data and materials

All data generated or analyzed during this study are included in this published article.

Authors' contributions

MRa contributed to conception and design, responsible for supervision, funding acquisition and writing the original draft of the manuscript. CS was responsible for supervision and writing the original draft of the manuscript and performed the formal analysis. MV was responsible for supervision and performign formal analysis. AnC, AlC, MC, RL, RP and AG were responsible for investigation, writing, reviewing and editing; ER, MRi and EP were responsible for formal analysis and data curation. FG and DM were responsible for formal analysis, writing, reviewing and editing. MRa and CS confirm the authenticity of all the raw data. All authors read and approved the final manuscript.

Ethics approval and consent to participate

The present study was conducted in accordance with the Declaration of Helsinki and approved by the Institutional Review Board (or Ethics Committee) of Sapienza University and Policlinico Umberto I, Rome, Italy; approval number: 6129. Informed consent was obtained from all subjects involved in the study.

Patient consent for publication

Not applicable.

Competing interests

The authors declare that they have no competing interests.

Glossary

Abbreviations

Abbreviations:

HNC

head and neck cancer

SCC

squamous cell carcinoma

LSCC

laryngeal squamous cell carcinoma

CGA

chromogranin A

NGF

nerve growth factor

proVGF

VGF precursor

FFPE

formalin-fixed paraffin-embedded

qPCR

quantitative real-time PCR

WB

western blotting

GAPDH

glyceraldehyde-3-phosphate dehydrogenase

CD3

cluster of differentiation 3

References

1 

Siegel RL, Miller KD and Jemal A: Cancer statistics, 2019. CA Cancer J Clin. 69:7–34. 2019. View Article : Google Scholar : PubMed/NCBI

2 

Johnson DE, Burtness B, Leemans CR, Lui VWY, Bauman JE and Grandis JR: Head and neck squamous cell carcinoma. Nat Rev Dis Prim. 6:922020. View Article : Google Scholar : PubMed/NCBI

3 

Bray F, Ferlay J, Soerjomataram I, Siegel RL, Torre LA and Jemal A: Global cancer statistics 2018: GLOBOCAN estimates of incidence and mortality worldwide for 36 cancers in 185 countries. CA Cancer J Clin. 68:394–424. 2018. View Article : Google Scholar : PubMed/NCBI

4 

Ferlito A, Silver CE, Bradford CR and Rinaldo A: Neuroendocrine neoplasms of the larynx: An overview. Head Neck. 31:1634–1646. 2009. View Article : Google Scholar : PubMed/NCBI

5 

Hunt JL, Barnes L, Triantafyllou A, Gnepp DR, Devaney KO, Stenman G, Halmos GB, Bishop JA, Skálová A, Willems SM, et al: Well-differentiated neuroendocrine carcinoma of the larynx: Confusion of terminology and uncertainty of early studies. Adv Anat Pathol. 26:246–250. 2019. View Article : Google Scholar : PubMed/NCBI

6 

Machiels JP, René Leemans C, Golusinski W, Grau C, Licitra L and Gregoire V; EHNS Executive Board, ESMO Guidelines Committee and ESTRO Executive Board, : Reprint of ‘Squamous cell carcinoma of the oral cavity, larynx, oropharynx and hypopharynx: EHNS-ESMO-ESTRO clinical practice guidelines for diagnosis, treatment and follow-up’. Oral Oncol. 113:1050422021. View Article : Google Scholar : PubMed/NCBI

7 

Forastiere AA, Ismaila N, Lewin JS, Nathan CA, Adelstein DJ, Eisbruch A, Fass G, Fisher SG, Laurie SA, Le QT, et al: Use of larynx-preservation strategies in the treatment of laryngeal cancer: American society of clinical oncology clinical practice guideline update. J Clin Oncol. 36:1143–1169. 2018. View Article : Google Scholar : PubMed/NCBI

8 

Portela-Gomes GM, Grimelius L, Wilander E and Stridsberg M: Granins and granin-related peptides in neuroendocrine tumours. Regul Pept. 165:12–20. 2010. View Article : Google Scholar : PubMed/NCBI

9 

Bartolomucci A, Possenti R, Mahata SK, Fischer-Colbrie R, Loh YP and Salton SRJ: The extended granin family: Structure, function, and biomedical implications. Endocr Rev. 32:755–797. 2011. View Article : Google Scholar : PubMed/NCBI

10 

Komiya A, Suzuki H, Imamoto T, Kamiya N, Nihei N, Naya Y, Ichikawa T and Fuse H: Neuroendocrine differentiation in the progression of prostate cancer. Int J Urol. 16:37–44. 2009. View Article : Google Scholar : PubMed/NCBI

11 

Massironi S, Conte D, Sciola V, Spampatti MP, Ciafardini C, Valenti L, Rossi RE and Peracchi M: Plasma chromogranin A response to octreotide test: Prognostic value for clinical outcome in endocrine digestive tumors. Am J Gastroenterol. 105:2072–2078. 2010. View Article : Google Scholar : PubMed/NCBI

12 

Kusafuka K, Abe M, Iida Y, Onitsuka T, Fuke T, Asano R, Kamijo T and Nakajima T: Mucosal large cell neuroendocrine carcinoma of the head and neck regions in Japanese patients: A distinct clinicopathological entity. J Clin Pathol. 65:704–709. 2012. View Article : Google Scholar : PubMed/NCBI

13 

Lewis JS Jr, Chernock RD and Bishop JA: Squamous and neuroendocrine specific immunohistochemical markers in head and neck squamous cell carcinoma: A tissue microarray study. Head Neck Pathol. 12:62–70. 2018. View Article : Google Scholar : PubMed/NCBI

14 

Wang KR, Jia YJ, Zhou SH, Wang QY, Bao YY, Feng ZY, Yao HT and Fan J: Cutaneous and subcutaneous metastases from atypical laryngeal carcinoids: Case report and review of the literature. Medicine (Baltimore). 95:e27962016. View Article : Google Scholar : PubMed/NCBI

15 

Salton SR, Ferri GL, Hahm S, Snyder SE, Wilson AJ, Possenti R and Levi A: VGF: A novel role for this neuronal and neuroendocrine polypeptide in the regulation of energy balance. Front Neuroendocrinol. 21:199–219. 2000. View Article : Google Scholar : PubMed/NCBI

16 

Bartolomucci A, La Corte G, Possenti R, Locatelli V, Rigamonti AE, Torsello A, Bresciani E, Bulgarelli I, Rizzi R, Pavone F, et al: TLQP-21, a VGF-derived peptide, increases energy expenditure and prevents the early phase of diet-induced obesity. Proc Natl Acad Sci USA. 103:14584–14589. 2006. View Article : Google Scholar : PubMed/NCBI

17 

Severini C, Ciotti MT, Biondini L, Quaresima S, Rinaldi AM, Levi A, Frank C and Possenti R: TLQP-21, a neuroendocrine VGF-derived peptide, prevents cerebellar granule cells death induced by serum and potassium deprivation. J Neurochem. 104:534–544. 2008. View Article : Google Scholar : PubMed/NCBI

18 

Shimazawa M, Tanaka H, Ito Y, Morimoto N, Tsuruma K, Kadokura M, Tamura S, Inoue T, Yamada M, Takahashi H, et al: An inducer of VGF protects cells against ER stress-induced cell death and prolongs survival in the mutant SOD1 animal models of familial ALS. PLoS One. 5:e153072010. View Article : Google Scholar : PubMed/NCBI

19 

Wang Y, Qin X, Han Y and Li B: VGF: A prospective biomarker and therapeutic target for neuroendocrine and nervous system disorders. Biomed Pharmacother. 151:1130992022. View Article : Google Scholar : PubMed/NCBI

20 

Annaratone L, Medico E, Rangel N, Castellano I, Marchiò C, Sapino A and Bussolati G: Search for neuro-endocrine markers (chromogranin A, synaptophysin and VGF) in breast cancers. An integrated approach using immunohistochemistry and gene expression profiling. Endocr Pathol. 25:219–228. 2014. View Article : Google Scholar : PubMed/NCBI

21 

Matsumoto T, Kawashima Y, Nagashio R, Kageyama T, Kodera Y, Jiang SX, Okayasu I, Kameya T and Sato Y: A new possible lung cancer marker: VGF detection from the conditioned medium of pulmonary large cell neuroendocrine carcinoma-derived cells using secretome analysis. Int J Biol Markers. 24:282–285. 2009. View Article : Google Scholar : PubMed/NCBI

22 

Ostrow KL, Park HL, Hoque MO, Kim MS, Liu J, Argani P, Westra W, Van Criekinge W and Sidransky D: Pharmacologic unmasking of epigenetically silenced genes in breast cancer. Clin Cancer Res. 15:1184–1191. 2009. View Article : Google Scholar : PubMed/NCBI

23 

Brait M, Maldonado L, Begum S, Loyo M, Wehle D, Tavora FF, Looijenga LHJ, Kowalski J, Zhang Z, Rosenbaum E, et al: DNA methylation profiles delineate epigenetic heterogeneity in seminoma and non-seminoma. Br J Cancer. 106:414–423. 2012. View Article : Google Scholar : PubMed/NCBI

24 

Brait M, Maldonado L, Noordhuis M, Begum S, Loyo M, Poeta ML, Barbosa A, Fazio VM, Angioli R, Rabitti C, et al: Association of promoter methylation of VGF and PGP9.5 with ovarian cancer progression. PLoS One. 8:e708782013. View Article : Google Scholar : PubMed/NCBI

25 

Nehar D, Lombard-Bohas C, Olivieri S, Claustrat B, Chayvialle JA, Penes MC, Sassolas G and Borson-Chazot F: Interest of chromogranin A for diagnosis and follow-up of endocrine tumours. Clin Endocrinol (Oxf). 60:644–652. 2004. View Article : Google Scholar : PubMed/NCBI

26 

Amin MB, Greene FL, Edge SB, Compton CC, Gershenwald JE, Brookland RK, Meyer L, Gress DM, Byrd DR and Winchester DP: The eighth edition AJCC cancer staging manual: Continuing to build a bridge from a population-based to a more ‘personalized’ approach to cancer staging. CA Cancer J Clin. 67:93–99. 2017. View Article : Google Scholar : PubMed/NCBI

27 

Livak KJ and Schmittgen TD: Analysis of relative gene expression data using real-time quantitative PCR and the 2(−Delta Delta C(T)) method. Methods. 25:402–408. 2001. View Article : Google Scholar : PubMed/NCBI

28 

Gabanella F, Colizza A, Mottola MC, Francati S, Blaconà G, Petrella C, Barbato C, Greco A, Ralli M, Fiore M, et al: The RNA-binding protein SMN as a novel player in laryngeal squamous cell carcinoma. Int J Mol Sci. 24:17942023. View Article : Google Scholar : PubMed/NCBI

29 

Zhou J, Tao D, Xu Q, Gao Z and Tang D: Expression of E-cadherin and vimentin in oral squamous cell carcinoma. Int J Clin Exp Pathol. 8:3150–3154. 2015.PubMed/NCBI

30 

Busse S, Steiner J, Micheel J, Dobrowolny H, Mawrin C, Krause TJ, Adamaszek M, Bogerts B, Bommhardt U, Hartig R and Busse M: Age-related increase of VGF-expression in T lymphocytes. Aging (Albany NY). 6:440–453. 2014. View Article : Google Scholar : PubMed/NCBI

31 

Busse S, Steiner J, Glorius S, Dobrowolny H, Greiner-Bohl S, Mawrin C, Bommhardt U, Hartig R, Bogerts B and Busse M: VGF expression by T lymphocytes in patients with Alzheimer's disease. Oncotarget. 6:14843–14851. 2015. View Article : Google Scholar : PubMed/NCBI

32 

Trani E, Giorgi A, Canu N, Amadoro G, Rinaldi AM, Halban PA, Ferri GL, Possenti R, Schininà ME and Levi A: Isolation and characterization of VGF peptides in rat brain. Role of PC1/3 and PC2 in the maturation of VGF precursor. J Neurochem. 81:565–574. 2002. View Article : Google Scholar : PubMed/NCBI

33 

Ferri GL, Noli B, Brancia C, D'Amato F and Cocco C: VGF: An inducible gene product, precursor of a diverse array of neuro-endocrine peptides and tissue-specific disease biomarkers. J Chem Neuroanat. 42:249–261. 2011. View Article : Google Scholar : PubMed/NCBI

34 

Bartolomucci A, Possenti R, Levi A, Pavone F and Moles A: The role of the vgf gene and VGF-derived peptides in nutrition and metabolism. Genes Nutr. 2:169–180. 2007. View Article : Google Scholar : PubMed/NCBI

35 

Rindi G, Licini L, Necchi V, Bottarelli L, Campanini N, Azzoni C, Favret M, Giordano G, D'Amato F, Brancia C, et al: Peptide products of the neurotrophin-inducible gene vgf are produced in human neuroendocrine cells from early development and increase in hyperplasia and neoplasia. J Clin Endocrinol Metab. 92:2811–2815. 2007. View Article : Google Scholar : PubMed/NCBI

36 

Hwang W, Chiu YF, Kuo MH, Lee KL, Lee AC, Yu CC, Chang JL, Huang WC, Hsiao SH, Lin SE and Chou YT: Expression of neuroendocrine factor VGF in lung cancer cells confers resistance to EGFR kinase inhibitors and triggers epithelial-to-mesenchymal transition. Cancer Res. 77:3013–3026. 2017. View Article : Google Scholar : PubMed/NCBI

37 

Hahm S, Mizuno TM, Wu TJ, Wisor JP, Priest CA, Kozak CA, Boozer CN, Peng B, McEvoy RC, Good P, et al: Targeted deletion of the Vgf gene indicates that the encoded secretory peptide precursor plays a novel role in the regulation of energy balance. Neuron. 23:537–548. 1999. View Article : Google Scholar : PubMed/NCBI

38 

Liu A and Curran MA: Tumor hypermetabolism confers resistance to immunotherapy. Semin Cancer Biol. 65:155–163. 2020. View Article : Google Scholar : PubMed/NCBI

39 

Chang CH, Qiu J, O'Sullivan D, Buck MD, Noguchi T, Curtis JD, Chen Q, Gindin M, Gubin MM, van der Windt GJ, et al: Metabolic competition in the tumor microenvironment is a driver of cancer progression. Cell. 162:1229–1241. 2015. View Article : Google Scholar : PubMed/NCBI

Related Articles

Journal Cover

January-2024
Volume 27 Issue 1

Print ISSN: 1792-1074
Online ISSN:1792-1082

Sign up for eToc alerts

Recommend to Library

Copy and paste a formatted citation
x
Spandidos Publications style
Gabanella F, Maftei D, Colizza A, Rullo E, Riminucci M, Pasqualucci E, Di Certo MG, Lattanzi R, Possenti R, Corsi A, Corsi A, et al: Reduced expression of secretogranin VGF in laryngeal squamous cell carcinoma. Oncol Lett 27: 37, 2024
APA
Gabanella, F., Maftei, D., Colizza, A., Rullo, E., Riminucci, M., Pasqualucci, E. ... Ralli, M. (2024). Reduced expression of secretogranin VGF in laryngeal squamous cell carcinoma. Oncology Letters, 27, 37. https://doi.org/10.3892/ol.2023.14170
MLA
Gabanella, F., Maftei, D., Colizza, A., Rullo, E., Riminucci, M., Pasqualucci, E., Di Certo, M. G., Lattanzi, R., Possenti, R., Corsi, A., Greco, A., De Vincentiis, M., Severini, C., Ralli, M."Reduced expression of secretogranin VGF in laryngeal squamous cell carcinoma". Oncology Letters 27.1 (2024): 37.
Chicago
Gabanella, F., Maftei, D., Colizza, A., Rullo, E., Riminucci, M., Pasqualucci, E., Di Certo, M. G., Lattanzi, R., Possenti, R., Corsi, A., Greco, A., De Vincentiis, M., Severini, C., Ralli, M."Reduced expression of secretogranin VGF in laryngeal squamous cell carcinoma". Oncology Letters 27, no. 1 (2024): 37. https://doi.org/10.3892/ol.2023.14170