Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 13.563
Filter
2.
Nat Commun ; 15(1): 4052, 2024 May 14.
Article in English | MEDLINE | ID: mdl-38744820

ABSTRACT

Obesity has emerged as a prominent risk factor for the development of malignant tumors. However, the existing literature on the role of adipocytes in the tumor microenvironment (TME) to elucidate the correlation between obesity and cancer remains insufficient. Here, we aim to investigate the formation of cancer-associated adipocytes (CAAs) and their contribution to tumor growth using mouse models harboring dysfunctional adipocytes. Specifically, we employ adipocyte-specific BECN1 KO (BaKO) mice, which exhibit lipodystrophy due to dysfunctional adipocytes. Our results reveal the activation of YAP/TAZ signaling in both CAAs and BECN1-deficient adipocytes, inducing adipocyte dedifferentiation and formation of a malignant TME. The additional deletion of YAP/TAZ from BaKO mice significantly restores the lipodystrophy and inflammatory phenotypes, leading to tumor regression. Furthermore, mice fed a high-fat diet (HFD) exhibit decreased BECN1 and increased YAP/TAZ expression in their adipose tissues. Treatment with the YAP/TAZ inhibitor, verteporfin, suppresses tumor progression in BaKO and HFD-fed mice, highlighting its efficacy against mice with metabolic dysregulation. Overall, our findings provide insights into the key mediators of CAA and their significance in developing a TME, thereby suggesting a viable approach targeting adipocyte homeostasis to suppress cancer growth.


Subject(s)
Adaptor Proteins, Signal Transducing , Adipocytes , Diet, High-Fat , Mice, Knockout , Tumor Microenvironment , YAP-Signaling Proteins , Animals , YAP-Signaling Proteins/metabolism , Adipocytes/metabolism , Adipocytes/pathology , Adaptor Proteins, Signal Transducing/metabolism , Adaptor Proteins, Signal Transducing/genetics , Mice , Diet, High-Fat/adverse effects , Transcription Factors/metabolism , Transcription Factors/genetics , Obesity/metabolism , Obesity/pathology , Humans , Verteporfin/pharmacology , Signal Transduction , Transcriptional Coactivator with PDZ-Binding Motif Proteins , Disease Progression , Male , Cell Transformation, Neoplastic/metabolism , Cell Transformation, Neoplastic/genetics , Cell Transformation, Neoplastic/pathology , Neoplasms/metabolism , Neoplasms/pathology , Neoplasms/genetics , Cell Cycle Proteins/metabolism , Cell Cycle Proteins/genetics , Lipodystrophy/metabolism , Lipodystrophy/pathology , Lipodystrophy/genetics , Mice, Inbred C57BL , Trans-Activators/metabolism , Trans-Activators/genetics
3.
BMJ Case Rep ; 17(5)2024 May 22.
Article in English | MEDLINE | ID: mdl-38782435

ABSTRACT

We present a novel case of a malignant transformation of an extremity soft tissue angioleiomyoma to leiomyosarcoma in a man in his late 70s who presented with a painful and increasing lump on his anterior tibia. Initial imaging and biopsy showed a benign angioleiomyoma which was excised for symptomatic reasons. An analysis of the resulting specimen revealed a 50×42×15 mm smooth muscle neoplasm consistent with angioleiomyoma with a 22×11 mm entirely intralesional nodular component in keeping with a grade 1 leiomyosarcoma. The malignant constituent of the lesion was entirely encased in benign angioleiomyoma negating the need for further surgery. Systemic staging investigation revealed no evidence of metastatic disease spread final staging as per the eighth edition of the American Joint Committee on Cancer (AJCC) Staging T1N0M0 R0 Stage 1 a.


Subject(s)
Angiomyoma , Leiomyosarcoma , Tibia , Humans , Male , Leiomyosarcoma/pathology , Leiomyosarcoma/surgery , Leiomyosarcoma/diagnostic imaging , Tibia/pathology , Tibia/diagnostic imaging , Angiomyoma/pathology , Angiomyoma/surgery , Angiomyoma/diagnostic imaging , Aged , Cell Transformation, Neoplastic/pathology , Soft Tissue Neoplasms/pathology , Soft Tissue Neoplasms/surgery , Soft Tissue Neoplasms/diagnostic imaging , Biopsy , Bone Neoplasms/pathology , Bone Neoplasms/surgery , Bone Neoplasms/diagnostic imaging
4.
Head Neck Pathol ; 18(1): 44, 2024 May 22.
Article in English | MEDLINE | ID: mdl-38775845

ABSTRACT

While acinic cell carcinoma (AciCC) can undergo high-grade transformation (HGT) to high-grade adenocarcinoma or poorly differentiated carcinoma, other morphologies such as spindle cell/sarcomatoid carcinoma are rare and not well-characterized. We herein report a novel case of AciCC with squamoglandular and chondrosarcomatous HGT mimicking a so-called 'carcinosarcoma ex-pleomorphic adenoma'. The patient is an 81-year-old male with a two-month history of neck swelling and referred otalgia who presented with a left parapharyngeal space mass extending into retropharyngeal space and pterygoid muscles. On resection, the tumor showed considerable morphologic diversity with high-grade serous and mucous acinar components as well as cribriform to solid apocrine-like components with comedonecrosis and squamous differentiation, all of which were embedded in a chondromyxoid background ranging from paucicellular and bland to a high-grade chondrosarcoma/pleomorphic sarcoma-like appearance. Only a minor conventional AciCC component was noted. Immunostains were negative for AR and only focally positive for GCDFP-15 arguing against a true apocrine phenotype, while PLAG1 and HMGA2 were negative arguing against an antecedent pleomorphic adenoma. On the other hand, SOX-10, DOG-1 and PAS after diastase highlighted serous acinar differentiation, and mucicarmine, and NKX3.1 highlighted mucous acinar differentiation. NR4A3 immunohistochemical staining and NR4A3 fluorescence in situ hybridization were positive in the carcinomatous and sarcomatoid components while sequencing analysis of both components revealed identical alterations involving TP53, PIK3CB, ARID1A, and STK11. This unique case warrants caution in designating all salivary sarcomatoid carcinomas with heterologous elements as part of the 'carcinoma ex-pleomorphic adenoma' family.


Subject(s)
Adenoma, Pleomorphic , Carcinoma, Acinar Cell , Salivary Gland Neoplasms , Humans , Male , Aged, 80 and over , Diagnosis, Differential , Carcinoma, Acinar Cell/pathology , Carcinoma, Acinar Cell/diagnosis , Adenoma, Pleomorphic/pathology , Adenoma, Pleomorphic/diagnosis , Salivary Gland Neoplasms/pathology , Salivary Gland Neoplasms/diagnosis , Carcinosarcoma/pathology , Cell Transformation, Neoplastic/pathology , Terminology as Topic , Chondrosarcoma/pathology , Chondrosarcoma/diagnosis
5.
BMC Oral Health ; 24(1): 588, 2024 May 21.
Article in English | MEDLINE | ID: mdl-38773401

ABSTRACT

BACKGROUND: White Sponge Nevus (WSN) is traditionally considered a benign genetic disorder affecting the oral mucosa, primarily caused by pathogenic mutations in keratin 4 (KRT4) or keratin 13 (KRT13). Despite its benign nature, recent evidence has begun to question the malignant potential of WSN. CASE PRESENTATION: We report a case involving a 70-year-old man who presented with a white lesion on the right floor of his mouth. Initial diagnostic evaluations confirmed the lesion as WSN. Over a one-year follow-up, the lesion underwent malignant transformation, evolving into local epithelial moderate-to-severe dysplasia. Exome sequencing identified a novel insertion mutation in exon 1 of the KRT4 gene, resulting in a deletion-insertion amino acid mutation involving glycine. Single-cell RNA sequencing further revealed altered epithelial proliferation and differentiation dynamics within the lesion. CONCLUSIONS: This case not only expands the known genetic spectrum of KRT4 mutations associated with WSN but also provides preliminary evidence suggesting the malignant potential of WSN. The novel pathogenic mutation in KRT4 is postulated to alter epithelial proliferation and differentiation, thereby raising concerns about the malignant transformation of WSN. Further studies are warranted to confirm these findings.


Subject(s)
Cell Transformation, Neoplastic , Keratin-4 , Leukokeratosis, Hereditary Mucosal , Humans , Male , Aged , Keratin-4/genetics , Cell Transformation, Neoplastic/genetics , Cell Transformation, Neoplastic/pathology , Leukokeratosis, Hereditary Mucosal/genetics , Leukokeratosis, Hereditary Mucosal/pathology , Mutation , Mouth Neoplasms/genetics , Mouth Neoplasms/pathology , Mouth Mucosa/pathology
6.
J Neuropathol Exp Neurol ; 83(6): 416-424, 2024 May 22.
Article in English | MEDLINE | ID: mdl-38699943

ABSTRACT

Ganglioglioma (GG) with anaplasia (anaplastic ganglioglioma) is a rare and controversial diagnosis. When present, anaplasia involves the glial component of the tumor, either at presentation or at recurrence. To date, most published cases lack molecular characterization. We describe the histologic and molecular features of 3 patients presenting with BRAF p. V600E-mutant GG (CNS WHO grade 1) with high-grade glial transformation at recurrence. The tumors occurred in pediatric patients (age 9-16 years) with time to recurrence from 20 months to 7 years. At presentation, each tumor was low-grade, with a BRAFV600E-positive ganglion cell component and a glial component resembling pleomorphic xanthoastrocytoma (PXA) or fibrillary astrocytoma. At recurrence, tumors resembled anaplastic PXA or high-grade astrocytomas without neuronal differentiation. CDKN2A homozygous deletion (HD) was absent in all primary tumors. At recurrence, 2 cases acquired CDKN2A HD; the third case showed loss of p16 and MTAP immunoexpression, but no CDKN2A/B HD or mutation was identified. By DNA methylation profiling, all primary and recurrent tumors either grouped or definitely matched to different methylation classes. Our findings indicate that malignant progression of the glial component can occur in GG and suggest that CDKN2A/B inactivation plays a significant role in this process.


Subject(s)
Brain Neoplasms , Ganglioglioma , Humans , Ganglioglioma/genetics , Ganglioglioma/pathology , Adolescent , Child , Brain Neoplasms/genetics , Brain Neoplasms/pathology , Male , Female , Proto-Oncogene Proteins B-raf/genetics , Epigenesis, Genetic , Neoplasm Recurrence, Local/genetics , Neoplasm Recurrence, Local/pathology , Cell Transformation, Neoplastic/genetics , Cell Transformation, Neoplastic/pathology
7.
BMC Oral Health ; 24(1): 639, 2024 May 30.
Article in English | MEDLINE | ID: mdl-38816724

ABSTRACT

BACKGROUND: Proliferative verrucous leukoplakia (PVL), distinguished by its malignant transformation rate of 43.87% to 65.8%, stands as the oral potentially malignant disorder with the highest propensity for malignancy. PVL is marked by distinctive heterogeneity regarding the clinical or histopathological characteristics as well as prognostic factors pertinent to this condition. The purpose of this study is to compile and assess the clinicopathological features, malignant transformation, and associated risk factors in patients diagnosed with PVL. METHODS: This study is a hospital-based retrospective longitudinal study of 36 patients diagnosed with PVL from 2013 to 2023. We conducted complete clinical and histopathological evaluations of the patients. RESULTS: The cohort comprised 16 males and 20 females, yielding a male-to-female ratio of 1:1.25. The follow-up period ranged from 8 to 125 months, with an average of 47.50 months. The most common clinical type of lesion was the verrucous form (58.33%), and the gingiva was the most common site (44.44%). Each patient had between 2 to 7 lesions, averaging 3.36 per patient. During the follow-up period, twelve patients (33.3%) developed oral cancer, with an average time to malignant transformation of 35.75 months. Kaplan-Meier survival analysis indicated that patients with complaints of pain, roughness, or a rough sensation, with diabetes, and the presence of cytologic atypia histologically showed a higher risk of malignant transformation (p < 0.05). In this study, the rate of malignant transformation in the treatment group (5/23) was lower than that in the untreated group (7/13), however, no statistically significant difference (p = 0.05). CONCLUSION: The main complaints of pain, roughness, or foreign body sensation, coupled with cytologic atypia histologically are indicative of an increased risk of malignant transformation in PVL. Further research is needed to elucidate the influence of these clinicopathological parameters on the malignant progression of PVL.


Subject(s)
Cell Transformation, Neoplastic , Leukoplakia, Oral , Humans , Male , Female , Leukoplakia, Oral/pathology , Cell Transformation, Neoplastic/pathology , Retrospective Studies , Middle Aged , Longitudinal Studies , Aged , Adult , Risk Factors , Mouth Neoplasms/pathology , Aged, 80 and over , Precancerous Conditions/pathology
8.
J Cancer Res Ther ; 20(2): 706-711, 2024 Apr 01.
Article in English | MEDLINE | ID: mdl-38687943

ABSTRACT

BACKGROUND: Oral submucous fibrosis (OSF) is a precancerous lesion, with oral squamous cell carcinoma (OSCC) being the most prevalent malignancy affecting the oral mucosa. The malignant transformation of OSF into OSCC is estimated to occur in 7-13% of cases. Myofibroblasts (MFs) play pivotal roles in both physiological and pathological processes, such as wound healing and tumorigenesis, respectively. This study aimed to explore the involvement of MFs in the progression of OSF and its malignant transformation. MATERIALS AND METHODS: In total, 94 formalin-fixed paraffin-embedded tissue blocks were collected, including normal oral mucosa (NOM; n = 10), early-moderate OSF (EMOSF; n = 29), advanced OSF (AOSF; n = 29), paracancerous OSF (POSF; n = 21), and OSCC (n = 5) samples. Alpha-smooth muscle actin was used for the immunohistochemical identification of MFs. RESULTS: NOM exhibited infrequent expression of MFs. A higher staining index of MFs was found in AOSF, followed by EMOSF and NOM. Additionally, a significant increase in the staining index of MFs was found from EMOSF to POSF and OSCC. The staining index of MFs in NOM, EMOSF, AOSF, POSF, and OSCC was 0.14 ± 0.2, 1.69 ± 1.4, 2.47 ± 1.2, 3.57 ± 2.6, and 8.86 ± 1.4, respectively. All results were statistically significant (P < 0.05). CONCLUSIONS: The expression of MFs exhibited a gradual increase as the disease progressed from mild to malignant transformation, indicating the contributory role of MFs in the fibrogenesis and potential tumorigenesis associated with OSF.


Subject(s)
Cell Transformation, Neoplastic , Immunohistochemistry , Mouth Neoplasms , Myofibroblasts , Oral Submucous Fibrosis , Humans , Oral Submucous Fibrosis/pathology , Oral Submucous Fibrosis/metabolism , Myofibroblasts/pathology , Myofibroblasts/metabolism , Cell Transformation, Neoplastic/pathology , Cell Transformation, Neoplastic/metabolism , Mouth Neoplasms/pathology , Mouth Neoplasms/metabolism , Male , Female , Mouth Mucosa/pathology , Mouth Mucosa/metabolism , Precancerous Conditions/pathology , Precancerous Conditions/metabolism , Middle Aged , Adult , Actins/metabolism , Disease Progression
9.
Ann Plast Surg ; 92(5): 575-579, 2024 May 01.
Article in English | MEDLINE | ID: mdl-38669586

ABSTRACT

ABSTRACT: Epidermal cysts are among the most common benign subcutaneous tumors. However, malignant transformation of benign epidermal cysts into squamous carcinomas has been reported. Owing to its low incidence rate, the clinical and pathological features of this condition are not well understood. This study aimed to analyze the clinical and pathological characteristics of the malignant transformation of epidermal cysts, which could suggest an appropriate treatment strategy. We conducted a retrospective study of 9 patients diagnosed with squamous cell carcinoma arising from epidermal cysts. All patients underwent surgical excision, and clinical information regarding patient demographics, tumor characteristics, treatment, and outcomes was analyzed. The average age at diagnosis was 57.3 years, with an average latency period of 15.4 years. Five patients had undergone prior cyst excision or drainage, with an average of 2.3 episodes of recurrence. Surgical excision was the primary treatment in all cases, and 2 patients with margin involvement at the final pathology underwent re-excision with additional resection margins. No recurrence was observed during the follow-up period. Four patients had immune dysregulation due to an underlying chronic kidney disease or cancer. Our study emphasizes the need for increased awareness of squamous cell carcinoma arising from epidermal cysts in patients with a history of cyst existence or recurrence, especially those with immune deficiencies. We expect these findings to contribute to early suspicion of malignant transformation and guide adequate clinical decision-making.


Subject(s)
Carcinoma, Squamous Cell , Epidermal Cyst , Skin Neoplasms , Adult , Aged , Female , Humans , Male , Middle Aged , Carcinoma, Squamous Cell/surgery , Carcinoma, Squamous Cell/pathology , Cell Transformation, Neoplastic/pathology , Epidermal Cyst/surgery , Epidermal Cyst/pathology , Retrospective Studies , Skin Neoplasms/pathology , Skin Neoplasms/surgery , Young Adult , Aged, 80 and over
10.
J Vet Intern Med ; 38(3): 1744-1750, 2024.
Article in English | MEDLINE | ID: mdl-38587203

ABSTRACT

Progressive carcinogenesis of a gastric polyp with transformation to gastric adenocarcinoma and subsequent development of leptomeningeal carcinomatosis is described in an adult male Scottish terrier. Presenting clinical signs consisted of vomiting with intermittent hematemesis. Surgical biopsies over the course of 14 months documented the progression from gastric polyp to minimally invasive gastric carcinoma to invasive gastric adenocarcinoma, a pathogenesis not previously documented in veterinary oncology. The patient ultimately developed neurologic pathology and was euthanized, and necropsy evaluation identified widespread carcinomatosis with accompanying leptomeningeal metastasis. As in humans, gastric polyps in dogs rarely have malignant potential.


Subject(s)
Adenocarcinoma , Dog Diseases , Meningeal Carcinomatosis , Stomach Neoplasms , Dogs , Animals , Dog Diseases/pathology , Stomach Neoplasms/veterinary , Stomach Neoplasms/pathology , Meningeal Carcinomatosis/veterinary , Meningeal Carcinomatosis/secondary , Meningeal Carcinomatosis/pathology , Male , Adenocarcinoma/veterinary , Adenocarcinoma/pathology , Adenocarcinoma/secondary , Cell Transformation, Neoplastic/pathology
11.
Cell Rep Med ; 5(5): 101510, 2024 May 21.
Article in English | MEDLINE | ID: mdl-38614093

ABSTRACT

Key gene mutations are essential for colorectal cancer (CRC) development; however, how the mutated tumor cells impact the surrounding normal cells to promote tumor progression has not been well defined. Here, we report that PIK3CA mutant tumor cells transmit oncogenic signals and result in malignant transformation of intestinal epithelial cells (IECs) via paracrine exosomal arachidonic acid (AA)-induced H3K4 trimethylation. Mechanistically, PIK3CA mutations sustain SGK3-FBW7-mediated stability of the cPLA2 protein, leading to the synthetic increase in AA, which is transported through exosome and accumulated in IECs. Transferred AA directly binds Menin and strengthens the interactions of Menin and MLL1/2 methyltransferase. Finally, the combination of VTP50469, an inhibitor of the Menin-MLL interaction, and alpelisib synergistically represses PDX tumors harboring PIK3CA mutations. Together, these findings unveil the metabolic link between PIK3CA mutant tumor cells and the IECs, highlighting AA as the potential target for the treatment of patients with CRC harboring PIK3CA mutations.


Subject(s)
Arachidonic Acid , Cell Transformation, Neoplastic , Chromatin Assembly and Disassembly , Class I Phosphatidylinositol 3-Kinases , Mutation , Class I Phosphatidylinositol 3-Kinases/genetics , Class I Phosphatidylinositol 3-Kinases/metabolism , Humans , Arachidonic Acid/metabolism , Animals , Mutation/genetics , Cell Transformation, Neoplastic/genetics , Cell Transformation, Neoplastic/metabolism , Cell Transformation, Neoplastic/pathology , Chromatin Assembly and Disassembly/genetics , Mice , Cell Line, Tumor , Colon/pathology , Colon/metabolism , Colorectal Neoplasms/genetics , Colorectal Neoplasms/pathology , Colorectal Neoplasms/metabolism , Exosomes/metabolism , Exosomes/genetics , Intestinal Mucosa/metabolism , Intestinal Mucosa/pathology , Histones/metabolism , Histones/genetics
12.
Anticancer Res ; 44(5): 1885-1894, 2024 May.
Article in English | MEDLINE | ID: mdl-38677721

ABSTRACT

BACKGROUND/AIM: Breast cancer is a leading cause of cancer-related deaths among women. Down-regulation of the tumor suppressor gene Cyld in breast cancer has been linked to a poor prognosis. This study investigated the role of Cyld in breast cancer using conditional mutant mouse models carrying a Cyld mutation, which inactivates the deubiquitinating activity of its protein product CYLD in mammary epithelial cells. MATERIALS AND METHODS: We examined the potential of CYLD inactivation to induce mammary tumors spontaneously or modify the susceptibility of mice to mammary tumorigenesis by DMBA treatment or ErbB2 over-expression. RESULTS: CYLD inactivation significantly increased susceptibility to breast cancer induced by either DMBA treatment or ErbB2 over-expression. Moreover, while CYLD inactivation alone did not lead to spontaneous mammary tumorigenesis, it did contribute to the formation of multifocal hyperplastic lesions in virgin mice of predominantly FVB/NJ background. CONCLUSION: Our study demonstrates the tumor enhancing potential of CYLD inactivation in mammary tumorigenesis in vivo and establishes novel relevant mouse models that can be exploited for developing prognostic and therapeutic protocols.


Subject(s)
Deubiquitinating Enzyme CYLD , Animals , Female , Mice , 9,10-Dimethyl-1,2-benzanthracene/toxicity , Breast Neoplasms/pathology , Breast Neoplasms/genetics , Breast Neoplasms/metabolism , Cell Transformation, Neoplastic/genetics , Cell Transformation, Neoplastic/metabolism , Cell Transformation, Neoplastic/pathology , Deubiquitinating Enzyme CYLD/genetics , Deubiquitinating Enzyme CYLD/metabolism , Mammary Neoplasms, Experimental/pathology , Mammary Neoplasms, Experimental/genetics , Mutation , Receptor, ErbB-2/genetics , Receptor, ErbB-2/metabolism , Tumor Suppressor Proteins/genetics , Tumor Suppressor Proteins/metabolism
13.
FEBS J ; 291(10): 2091-2093, 2024 May.
Article in English | MEDLINE | ID: mdl-38646863

ABSTRACT

Cellular immortalization is a complex process that requires multiple genetic alterations to overcome restricting barriers, including senescence. Not surprisingly, many of these alterations are associated with cancer; two tumor suppressor pathways, the cellular tumor antigen p53 and p16-Retinoblastoma (RB) pathways, are the best-characterized examples, but their mutations alone are known to be insufficient to drive full immortalization. En et al. identified a role for the lamin B receptor (LBR) in promoting cellular proliferation and immortalization in p53- and RB-deficient cells by maintaining their genome integrity and suppressing senescence. Thus, modulation of LBR could be exploited to treat cancer and potentially also to promote cell rejuvenation.


Subject(s)
Cellular Senescence , Genomic Instability , Lamin B Receptor , Tumor Suppressor Protein p53 , Cellular Senescence/genetics , Humans , Tumor Suppressor Protein p53/genetics , Tumor Suppressor Protein p53/metabolism , Receptors, Cytoplasmic and Nuclear/genetics , Receptors, Cytoplasmic and Nuclear/metabolism , Animals , Retinoblastoma Protein/genetics , Retinoblastoma Protein/metabolism , Neoplasms/genetics , Neoplasms/pathology , Neoplasms/metabolism , Cell Proliferation/genetics , Cell Transformation, Neoplastic/genetics , Cell Transformation, Neoplastic/metabolism , Cell Transformation, Neoplastic/pathology
14.
Am J Surg Pathol ; 48(6): 652-661, 2024 Jun 01.
Article in English | MEDLINE | ID: mdl-38584451

ABSTRACT

Very well-differentiated adenocarcinoma of intestinal type is a distinct subtype of gastric cancer characterized by anastomosing glands with a hand-in-hand pattern and low-grade cytologic atypia resembling intestinal metaplasia. This is a slow-growing neoplasm with an indolent clinical course; however, a subset demonstrates transformation into adenocarcinoma with higher-grade histology, typically diffuse-type carcinoma, and behaves aggressively. This study aimed to better characterize the genomic and pathologic features, with a focus on factors associated with diffuse-type transformation. A total of 58 cases with (n=31) and without (n=27) diffuse-type transformation were analyzed for molecular and pathologic features. First, comprehensive deep DNA sequencing was conducted in 18 cases (discovery cohort), followed by a digital droplet polymerase chain reaction of hot spot RHOA mutations in 40 cases (validation cohort). In total, RHOA mutations were the most common alteration (34%), followed by loss of ARID1A (12%), p53 alterations (10%), and CLDN18 :: ARHGAP26/6 fusions (3.4%). FGFR2 amplification was identified in an advanced case with a p53 alteration. Altered p53 expression was recognized only in higher-grade components and was significantly associated with advanced disease ( P =0.0015) and diffuse-type transformation ( P =0.026). A mixed mucin phenotype was also strongly correlated with advanced disease ( P <0.001) and diffuse-type transformation ( P <0.001). Decreased E-cadherin expression was frequently observed (74%) in poorly cohesive components. This study demonstrated that a subset of RHOA -mutant diffuse-type gastric cancers develops through the transformation of very well-differentiated adenocarcinoma of intestinal type. Our observations suggest a mixed mucin phenotype as a risk factor and alterations in p53 and E-cadherin as drivers of diffuse-type transformation.


Subject(s)
Adenocarcinoma , Biomarkers, Tumor , Cell Transformation, Neoplastic , Mutation , Stomach Neoplasms , Humans , Stomach Neoplasms/genetics , Stomach Neoplasms/pathology , Adenocarcinoma/genetics , Adenocarcinoma/pathology , Adenocarcinoma/chemistry , Male , Female , Middle Aged , Aged , Cell Transformation, Neoplastic/genetics , Cell Transformation, Neoplastic/pathology , Biomarkers, Tumor/genetics , Biomarkers, Tumor/analysis , rhoA GTP-Binding Protein/genetics , Cell Differentiation , Adult , Phenotype , Aged, 80 and over , Tumor Suppressor Protein p53/genetics , Genetic Predisposition to Disease , DNA Mutational Analysis , High-Throughput Nucleotide Sequencing
15.
Nature ; 629(8011): 450-457, 2024 May.
Article in English | MEDLINE | ID: mdl-38658753

ABSTRACT

Three-dimensional organoid culture technologies have revolutionized cancer research by allowing for more realistic and scalable reproductions of both tumour and microenvironmental structures1-3. This has enabled better modelling of low-complexity cancer cell behaviours that occur over relatively short periods of time4. However, available organoid systems do not capture the intricate evolutionary process of cancer development in terms of tissue architecture, cell diversity, homeostasis and lifespan. As a consequence, oncogenesis and tumour formation studies are not possible in vitro and instead require the extensive use of animal models, which provide limited spatiotemporal resolution of cellular dynamics and come at a considerable cost in terms of resources and animal lives. Here we developed topobiologically complex mini-colons that are able to undergo tumorigenesis ex vivo by integrating microfabrication, optogenetic and tissue engineering approaches. With this system, tumorigenic transformation can be spatiotemporally controlled by directing oncogenic activation through blue-light exposure, and emergent colon tumours can be tracked in real-time at the single-cell resolution for several weeks without breaking the culture. These induced mini-colons display rich intratumoural and intertumoural diversity and recapitulate key pathophysiological hallmarks displayed by colorectal tumours in vivo. By fine-tuning cell-intrinsic and cell-extrinsic parameters, mini-colons can be used to identify tumorigenic determinants and pharmacological opportunities. As a whole, our study paves the way for cancer initiation research outside living organisms.


Subject(s)
Cell Transformation, Neoplastic , Colon , Colorectal Neoplasms , Optogenetics , Organoids , Animals , Humans , Mice , Cell Transformation, Neoplastic/pathology , Cell Transformation, Neoplastic/radiation effects , Colon/pathology , Colon/radiation effects , Colorectal Neoplasms/etiology , Colorectal Neoplasms/pathology , Light , Optogenetics/methods , Organoids/pathology , Organoids/radiation effects , Single-Cell Analysis , Time Factors , Tissue Engineering/methods , Tumor Microenvironment , Drug Evaluation, Preclinical
16.
Eur J Surg Oncol ; 50(6): 108247, 2024 Jun.
Article in English | MEDLINE | ID: mdl-38522332

ABSTRACT

INTRODUCTION: Endometriosis affects 10% of women of reproductive age. There is evidence for a left lateral predisposition of endometriotic lesions and a 1.9-fold greater risk of ovarian cancer in endometriosis. The aim of this study is to determine whether a left lateral predisposition of ovarian clear-cell carcinoma (CCC) and endometrioid carcinoma (EC) exists. MATERIALS AND METHODS: A retrospective cohort study of all EC and CCC patients in Northern Ireland between March-2011 and June-2018. ANOVA was used to analyse preoperative prediction of stage, chi-squared (χ2) was used to compare left- and right-sided masses. Survival was estimated using Kaplan-Meier and log-rank test. A p-value <0.05 was considered significant. RESULTS: 158 patients were identified (95 EC, 55 CCC, 8 mixed). Mean age was 57.65 years with 69% presenting at stage 1. The mean CA125 was 559 U/mL (p = 0.850) and mean abdominal mass size was 14.12 cm (p = 0.732). The most common presenting symptom was an abdominal mass (37%). Despite 67% of patients having endometriosis on final pathology, only 8.9% had a known history pre-operatively. 51% of tumours were located on the left (p = 0.036). For unilateral tumours this was significant for EC (P = 0.002) but not for CCC (P = 0.555). The 1-, 3- and 5-year overall survival for all types/stages was 85%, 78% and 71% respectively. CONCLUSION: While CCC and EC are associated with endometriosis, only EC exhibits a left lateral predisposition. There is no association between preoperative CA125 or abdominal mass size and stage of disease.


Subject(s)
Adenocarcinoma, Clear Cell , Carcinoma, Endometrioid , Cell Transformation, Neoplastic , Endometriosis , Ovarian Neoplasms , Humans , Female , Endometriosis/pathology , Endometriosis/complications , Carcinoma, Endometrioid/pathology , Ovarian Neoplasms/pathology , Middle Aged , Retrospective Studies , Adenocarcinoma, Clear Cell/pathology , Cell Transformation, Neoplastic/pathology , Adult , CA-125 Antigen/blood , Aged , Neoplasm Staging , Northern Ireland/epidemiology , Survival Rate
17.
Head Neck ; 46(5): 985-1000, 2024 May.
Article in English | MEDLINE | ID: mdl-38482546

ABSTRACT

OBJECTIVE: This study used array comparative genomic hybridization to assess copy number alterations (CNAs) involving miRNA genes in pleomorphic adenoma (PA), recurrent pleomorphic adenoma (RPA), residual PA, and carcinoma ex pleomorphic adenoma (CXPA). MATERIALS AND METHODS: We analyzed 13 PA, 4 RPA, 29 CXPA, and 14 residual PA using Nexus Copy Number Discovery software. The miRNAs genes affected by CNAs were evaluated based on their expression patterns and subjected to pathway enrichment analysis. RESULTS: Across the groups, we found 216 CNAs affecting 2261 miRNA genes, with 117 in PA, 59 in RPA, 846 in residual PA, and 2555 in CXPA. The chromosome 8 showed higher involvement in altered miRNAs in PAs and CXPA patients. Six miRNA genes were shared among all groups. Additionally, miR-21, miR-455-3p, miR-140, miR-320a, miR-383, miR-598, and miR-486 were prominent CNAs found and is implicated in carcinogenesis of several malignant tumors. These miRNAs regulate critical signaling pathways such as aerobic glycolysis, fatty acid biosynthesis, and cancer-related pathways. CONCLUSION: This study was the first to explore CNAs in miRNA-encoding genes in the PA-CXPA sequence. The findings suggest the involvement of numerous miRNA genes in CXPA development and progression by regulating oncogenic signaling pathways.


Subject(s)
Adenocarcinoma , Adenoma, Pleomorphic , MicroRNAs , Salivary Gland Neoplasms , Humans , Adenoma, Pleomorphic/genetics , Adenoma, Pleomorphic/pathology , DNA Copy Number Variations , Salivary Gland Neoplasms/pathology , MicroRNAs/genetics , Comparative Genomic Hybridization , Cell Transformation, Neoplastic/pathology , Adenocarcinoma/pathology
19.
Biochim Biophys Acta Mol Basis Dis ; 1870(5): 167133, 2024 Jun.
Article in English | MEDLINE | ID: mdl-38531482

ABSTRACT

The cytosolic dipeptidyl-aminopeptidase 9 (DPP9) cleaves protein N-termini post-proline or -alanine. Our analysis of DPP9 mRNA expression from the TCGA 'breast cancer' data set revealed that low/intermediate DPP9 levels are associated with poor overall survival of breast cancer patients. To unravel the impact of DPP9 on breast cancer development and progression, the transgenic MMTV-PyMT mouse model of metastasizing breast cancer was used. In addition, tissue- and time-controlled genetic deletion of DPP9 by the Cre-loxP recombination system was done. Despite a delay of tumor onset, a higher number of lung metastases were measured in DPP9-deficient mice compared to controls. In human mammary epithelial cells with oncogenic RAS pathway activation, DPP9 deficiency delayed tumorigenic transformation and accelerated TGF-ß1 induced epithelial-to-mesenchymal transition (EMT) of spheroids. For further analysis of the mechanism, primary breast tumor cells were isolated from the MMTV-PyMT model. DPP9 deficiency in these cells caused cancer cell migration and invasion accompanied by EMT. In absence of DPP9, the EMT transcription factor ZEB1 was stabilized due to insufficient degradation by the proteasome. In summary, low expression of DPP9 appears to decelerate mammary tumorigenesis but favors EMT and metastasis, which establishes DPP9 as a novel dynamic regulator of breast cancer initiation and progression.


Subject(s)
Breast Neoplasms , Dipeptidyl-Peptidases and Tripeptidyl-Peptidases , Epithelial-Mesenchymal Transition , Animals , Humans , Female , Dipeptidyl-Peptidases and Tripeptidyl-Peptidases/metabolism , Dipeptidyl-Peptidases and Tripeptidyl-Peptidases/genetics , Mice , Breast Neoplasms/pathology , Breast Neoplasms/genetics , Breast Neoplasms/metabolism , Carcinogenesis/genetics , Carcinogenesis/metabolism , Carcinogenesis/pathology , Lung Neoplasms/pathology , Lung Neoplasms/genetics , Lung Neoplasms/secondary , Lung Neoplasms/metabolism , Neoplasm Metastasis , Gene Expression Regulation, Neoplastic , Cell Transformation, Neoplastic/genetics , Cell Transformation, Neoplastic/metabolism , Cell Transformation, Neoplastic/pathology , Cell Line, Tumor , Mice, Knockout , Mice, Transgenic
20.
Blood ; 143(22): 2245-2255, 2024 May 30.
Article in English | MEDLINE | ID: mdl-38498034

ABSTRACT

ABSTRACT: The spectrum of myeloid disorders ranges from aplastic bone marrow failure characterized by an empty bone marrow completely lacking in hematopoiesis to acute myeloid leukemia in which the marrow space is replaced by undifferentiated leukemic blasts. Recent advances in the capacity to sequence bulk tumor population as well as at a single-cell level has provided significant insight into the stepwise process of transformation to acute myeloid leukemia. Using models of progression in the context of germ line predisposition (trisomy 21, GATA2 deficiency, and SAMD9/9L syndrome), premalignant states (clonal hematopoiesis and clonal cytopenia of unknown significance), and myelodysplastic syndrome, we review the mechanisms of progression focusing on the hierarchy of clonal mutation and potential roles of transcription factor alterations, splicing factor mutations, and the bone marrow environment in progression to acute myeloid leukemia. Despite major advances in our understanding, preventing the progression of these disorders or treating them at the acute leukemia phase remains a major area of unmet medical need.


Subject(s)
Disease Progression , Humans , Preleukemia/pathology , Preleukemia/genetics , Myelodysplastic Syndromes/pathology , Myelodysplastic Syndromes/genetics , Leukemia, Myeloid, Acute/pathology , Leukemia, Myeloid, Acute/genetics , Animals , Precancerous Conditions/pathology , Precancerous Conditions/genetics , Mutation , Cell Transformation, Neoplastic/genetics , Cell Transformation, Neoplastic/pathology , Cell Transformation, Neoplastic/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...