The influence of chemotherapy on the distribution of 57Co-bleomycin in chemically induced squamous cell carcinoma of mouse skin

1979 ◽  
Vol 4 (1) ◽  
Author(s):  
T. Pertynski ◽  
G. Reznik ◽  
P. Mari� ◽  
M.B. Ketkar ◽  
H. Haindl ◽  
...  
Author(s):  
Weronika Szukala ◽  
Agata Lichawska-Cieslar ◽  
Roza Pietrzycka ◽  
Maria Kulecka ◽  
Izabela Rumienczyk ◽  
...  

Abstract Background Squamous cell carcinoma (SCC) of the skin is a common form of nonmelanoma skin cancer. Monocyte chemotactic protein 1-induced protein 1 (MCPIP1), also called Regnase-1, is an RNase with anti-inflammatory properties. In normal human skin, its expression is predominantly restricted to the suprabasal epidermis. The main aim of this study was to investigate whether MCPIP1 is involved in the pathogenesis of SCC. Methods We analyzed the distribution of MCPIP1 in skin biopsies of patients with actinic keratoses (AKs) and SCCs. To explore the mechanisms by which MCPIP1 may modulate tumorigenesis in vivo, we established a mouse model of chemically induced carcinogenesis. Results Skin expression of MCPIP1 changed during the transformation of precancerous lesions into cutaneous SCC. MCPIP1 immunoreactivity was high in the thickened area of the AK epidermis but was predominantly restricted to keratin pearls in fully developed SCC lesions. Accelerated development of chemically induced skin tumors was observed in mice with loss of epidermal MCPIP1 (Mcpip1eKO). Papillomas that developed in Mcpip1eKO mouse skin were larger and characterized by elevated expression of markers typical of keratinocyte proliferation and tumor angiogenesis. This phenotype was correlated with enhanced expression of IL-6, IL-33 and transforming growth factor-beta (TGF-β). Moreover, our results demonstrated that in keratinocytes, the RNase MCPIP1 is essential for the negative regulation of genes encoding SCC antigens and matrix metallopeptidase 9. Conclusions Overall, our results provide a mechanistic understanding of how MCPIP1 contributes to the development of epidermoid carcinoma.


2016 ◽  
Vol 22 (2) ◽  
pp. 217-217 ◽  
Author(s):  
Dany Nassar ◽  
Mathilde Latil ◽  
Bram Boeckx ◽  
Diether Lambrechts ◽  
Cédric Blanpain

2011 ◽  
Vol 51 (9) ◽  
pp. 1717-1726 ◽  
Author(s):  
Halina Was ◽  
Malgorzata Sokolowska ◽  
Aleksandra Sierpniowska ◽  
Paweł Dominik ◽  
Klaudia Skrzypek ◽  
...  

2015 ◽  
Vol 10 (4) ◽  
pp. 2432-2438 ◽  
Author(s):  
DANIEL DROGUETT ◽  
CHRISTIAN CASTILLO ◽  
ELBA LEIVA ◽  
CRISTINA THEODULOZ ◽  
GUILLERMO SCHMEDA-HIRSCHMANN ◽  
...  

2017 ◽  
Vol 49 (11) ◽  
pp. 1624-1632 ◽  
Author(s):  
Phillips Y Huang ◽  
Eve Kandyba ◽  
Arnaud Jabouille ◽  
Jonas Sjolund ◽  
Atul Kumar ◽  
...  

Planta Medica ◽  
2015 ◽  
Vol 81 (16) ◽  
Author(s):  
W Shebaby ◽  
G Daher ◽  
R Taleb ◽  
H Daaboul ◽  
M El Sibai ◽  
...  

2004 ◽  
Vol 64 (5) ◽  
pp. 1647-1654 ◽  
Author(s):  
Yian Wang ◽  
Zhongqiu Zhang ◽  
Ying Yan ◽  
William J. Lemon ◽  
Marie LaRegina ◽  
...  

Oncogene ◽  
2021 ◽  
Author(s):  
Shukai Yuan ◽  
Peitao Zhang ◽  
Liqi Wen ◽  
Shikai Jia ◽  
Yufan Wu ◽  
...  

AbstractEmerging evidence suggests that the cancer stem cells (CSCs) are key culprits of cancer metastasis and drug resistance. Understanding mechanisms regulating the critical oncogenic pathways and CSCs function could reveal new diagnostic and therapeutic strategies. We now report that miR-22, a miRNA critical for hair follicle stem/progenitor cell differentiation, promotes tumor initiation, progression, and metastasis by maintaining Wnt/β-catenin signaling and CSCs function. Mechanistically, we find that miR-22 facilitates β-catenin stabilization through directly repressing citrullinase PAD2. Moreover, miR-22 also relieves DKK1-mediated repression of Wnt/β-catenin signaling by targeting a FosB-DDK1 transcriptional axis. miR-22 knockout mice showed attenuated Wnt/β-catenin activity and Lgr5+ CSCs penetrance, resulting in reduced occurrence, progression, and metastasis of chemically induced cutaneous squamous cell carcinoma (cSCC). Clinically, miR-22 is abundantly expressed in human cSCC. Its expression is even further elevated in the CSCs proportion, which negatively correlates with PAD2 and FosB expression. Inhibition of miR-22 markedly suppressed cSCC progression and increased chemotherapy sensitivity in vitro and in xenograft mice. Together, our results revealed a novel miR-22-WNT-CSCs regulatory mechanism in cSCC and highlight the important clinical application prospects of miR-22, a common target molecule for Wnt/β-catenin signaling and CSCs, for patient stratification and therapeutic intervention.


Sign in / Sign up

Export Citation Format

Share Document