scispace - formally typeset
Open AccessJournal ArticleDOI

A Rac1/Cdc42 GTPase-specific small molecule inhibitor suppresses growth of primary human prostate cancer xenografts and prolongs survival in mice.

Reads0
Chats0
TLDR
A role of AZA1 in blocking Rac1/Cdc42-dependent cell cycle progression, cancer cell migration and increase of cancer cell apoptosis involving down-regulation of the AKT and PAK signaling pathway in prostate cancer cells is suggested.
Abstract
Deregulated Rho GTPases Rac1 and Cdc42 have been discovered in various tumors, including prostate and Rac protein expression significantly increases in prostate cancer. The Rac and Cdc42 pathways promote the uncontrolled proliferation, invasion and metastatic properties of human cancer cells. We synthesized the novel compound AZA1 based on structural information of the known Rac1 inhibitor NSC23766. In the current study we investigated the effects of inhibition of these pathways by AZA1 on prostate tumorigenicity by performing preclinical studies using a xenograft mouse model of prostate cancer. In androgen-independent prostate cancer cells, AZA1 inhibited both Rac1 and Cdc42 but not RhoA GTPase activity in a dose-dependent manner and blocked cellular migration and proliferation. Cyclin D1 expression significantly decreased following Rac1/Cdc42 inhibition in prostate cancer cells. AZA1 treatment also down-regulated PAK and AKT activity in prostate cancer cells, associated with induction of the pro-apoptotic function of BAD by suppression of serine-112 phosphorylation. Daily systemic administration of AZA1 for 2 weeks reduced growth of human 22Rv1 prostate tumor xenografts in mice and improved the survival of tumor-bearing animals significantly. These data suggest a role of AZA1 in blocking Rac1/Cdc42-dependent cell cycle progression, cancer cell migration and increase of cancer cell apoptosis involving down-regulation of the AKT and PAK signaling pathway in prostate cancer cells. We therefore propose that a small-molecule inhibitor therapy targeting Rac1/Cdc42 Rho GTPase signaling pathways may be used as a novel treatment for patients with advanced prostate cancer.

read more

Content maybe subject to copyright    Report

Citations
More filters
Journal ArticleDOI

Targeting Rac and Cdc42 GTPases in Cancer.

TL;DR: The regulatory mechanisms, inhibitory efficacy, and the anticancer potential of Rac- and Cdc42-targeting agents are summarized and an understanding of the regulatory mechanisms of these pivotal signaling intermediates is key for the development of effective inhibitors.
Journal ArticleDOI

Deregulation of Rho GTPases in cancer

TL;DR: This review explores the various ways in which Rho signaling can be deregulated in human cancers and suggests that targeting Rho GTPase signaling could constitute an effective treatment.
Journal ArticleDOI

Rho GTPases modulate malignant transformation of tumor cells

TL;DR: How Rho GTPases contribute to other hallmarks of cancer with a special emphasis on malignant transformation is reviewed.
Journal ArticleDOI

Paving the Rho in cancer metastasis: Rho GTPases and beyond.

TL;DR: This review focuses on key discoveries in the regulation of epithelial-mesenchymal-transition, cell-cell junctions, formation of membrane protrusions, plasticity of cell migration and adaptation to a hypoxic environment and on crosstalk between Rho GTPase family members and other important oncogenic pathways.
Journal ArticleDOI

Approaches of targeting Rho GTPases in cancer drug discovery

TL;DR: This review focuses on the recent progress in targeting the signaling activities of three prototypical Rho GTPases, that is, RhoA, Rac1, and Cdc42, that are key regulators and effectors in cancer.
References
More filters
Journal ArticleDOI

Cancer Statistics, 2009

TL;DR: The most recent data on cancer incidence, mortality, and survival from the American Cancer Society (ACS) is presented in this paper, where the authors compare the three major cancer sites in men (lung, prostate, and colon and rectum [colorectum]) and in two major cancers sites in women (breast and colorectal) over a 15-year period.
Journal ArticleDOI

Akt Phosphorylation of BAD Couples Survival Signals to the Cell-Intrinsic Death Machinery

TL;DR: It is shown that growth factor activation of the PI3'K/Akt signaling pathway culminates in the phosphorylation of the BCL-2 family member BAD, thereby suppressing apoptosis and promoting cell survival.
Journal ArticleDOI

Opposing Effects of ERK and JNK-p38 MAP Kinases on Apoptosis

TL;DR: The effects of dominant-interfering or constitutively activated forms of various components of the JNK-p38 and ERK signaling pathways demonstrated that activation of JNK and p38 and concurrent inhibition of ERK are critical for induction of apoptosis in these cells.
Journal ArticleDOI

Docetaxel plus Prednisone or Mitoxantrone plus Prednisone for Advanced Prostate Cancer

TL;DR: When given with prednisone, treatment with docetaxel every three weeks led to superior survival and improved rates of response in terms of pain, serum PSA level, and quality of life, as compared with mitoxantrone plusprednisone.
Journal ArticleDOI

Cellular Motility Driven by Assembly and Disassembly of Actin Filaments

TL;DR: A core set of proteins including actin, Arp2/3 complex, profilin, capping protein, and ADF/cofilin can reconstitute the process in vitro, and mathematical models of the constituent reactions predict the rate of motion.
Related Papers (5)