Abstract

Mechanisms underlying sex differences in cancer incidence are not defined but likely involve dimorphism (s) in tumor suppressor function at the cellular and organismal levels. As an example, sexual dimorphism in retinoblastoma protein (Rb) activity was shown to block transformation of female, but not male, murine astrocytes in which neurofibromin and p53 function was abrogated (GBM astrocytes). Correlated sex differences in gene expression in the murine GBM astrocytes were found to be highly concordant with sex differences in gene expression in male and female GBM patients, including in the expression of components of the Rb and p53 pathways. To define the basis of this phenomenon, we examined the functions of the cyclin dependent kinase (CDK) inhibitors, p16, p21 and p27 in murine GBM astrocytes under conditions that promote Rb-dependent growth arrest. We found that upon serum deprivation or etoposide-induced DNA damage, female, but not male GBM astrocytes, respond with increased p16 and p21 activity, and cell cycle arrest. In contrast, male GBM astrocytes continue to proliferate, accumulate chromosomal aberrations, exhibit enhanced clonogenic cell activity and in vivo tumorigenesis; all manifestations of broad sex differences in cell cycle regulation and DNA repair. Differences in tumorigenesis disappeared when female GBM astrocytes are also rendered null for p16 and p21. These data elucidate mechanisms underlying sex differences in cancer incidence and demonstrate sex-specific effects of cytotoxic and targeted therapeutics. This has critical implications for lab and clinical research.

Highlights

  • Increasing volumes of data indicate significant sex differences exist in many human diseases [10, 14, 20, 24, 29, 31]

  • We measured in vivo tumorigenesis in male and female Cas9-expressing CD-1 IGS mice following in utero electroporation of guide RNAs (gRNAs) targeting the Nf1 and p53 genes into peri-ventricular neural progenitors

  • Increased necrosis has been reported to be a more prominent feature in female patient pathological specimens and magnetic resonance imaging [9, 12]. These data indicate that sex differences in tumorigenesis after combined loss of neurofibromin and p53 function are evident in different mouse strains and regardless of whether the loss is engineered in vitro or in vivo and independent of the method by which p53 function is abrogated

Read more

Summary

Introduction

Increasing volumes of data indicate significant sex differences exist in many human diseases [10, 14, 20, 24, 29, 31]. Sex differences in incidence presumably reflect differences in the mechanisms that determine vulnerability to disease [15, 25, 33, 35, 37]. Most notably those cancers that are sex hormone responsive, greater male prevalence is true regardless of age, cancer type, Multiple factors can contribute to sex differences in human health and disease. Sex differences arise through the multiple mechanisms of sexual differentiation. Starting with differences in sex chromosome complement and sex specific reprogramming of imprinted loci, and continuing with the organizational or epigenetic effects of in utero and pubertal sex hormones, and maturing with the acute action of Kfoury et al Acta Neuropathologica Communications (2018) 6:12 circulating sex hormones, sex differences are powerfully ensconced at the cellular and organismal levels

Objectives
Methods
Results
Conclusion
Full Text
Published version (Free)

Talk to us

Join us for a 30 min session where you can share your feedback and ask us any queries you have

Schedule a call