Abstract

Cancer-associated thrombosis is the second-leading cause of mortality in patients with cancer and presents a poor prognosis, with a lack of effective treatment strategies. NAD(P)H quinone oxidoreductase 1 (NQO1) increases the cellular nicotinamide adenine dinucleotide (NAD+) levels by accelerating the oxidation of NADH to NAD+, thus playing important roles in cellular homeostasis, energy metabolism, and inflammatory responses. Using a murine orthotopic 4T1 breast cancer model, in which multiple thrombi are generated in the lungs at the late stage of cancer development, we investigated the effects of regulating the cellular NAD+ levels on cancer-associated thrombosis. In this study, we show that dunnione (a strong substrate of NQO1) attenuates the prothrombotic state and lung thrombosis in tumor-bearing mice by inhibiting the expression of tissue factor and formation of neutrophil extracellular traps (NETs). Dunnione increases the cellular NAD+ levels in lung tissues of tumor-bearing mice to restore the declining sirtuin 1 (SIRT1) activity, thus deacetylating nuclear factor-kappa B (NF-κB) and preventing the overexpression of tissue factor in bronchial epithelial and vascular endothelial cells. In addition, we demonstrated that dunnione abolishes the ability of neutrophils to generate NETs by suppressing histone acetylation and NADPH oxidase (NOX) activity. Overall, our results reveal that the regulation of cellular NAD+ levels by pharmacological agents may inhibit pulmonary embolism in tumor-bearing mice, which may potentially be used as a viable therapeutic approach for the treatment of cancer-associated thrombosis.

Highlights

  • Cancer is commonly associated with hypercoagulability and a high risk of thrombosis, which make it the second leading cause of mortality in patients with cancer [1]

  • These results demonstrate that dunnione inhibits pulmonary thrombosis in advanced 4T1 breast cancer and suggests a pro-thrombotic role of neutrophils in 4T1 breast cancer mice

  • Emerging evidence indicates that neutrophils and neutrophil extracellular traps play essential roles in thrombogenesis, especially in cancer-associated thrombosis, because neutrophils are frequently remodeled with an increasing amount and are more prone to generate NETs in cancers [12,13,14,15,33]. 4T1, a breast cancer cell line that is usually used in animal models to mimic stage IV human breast cancer, has recently been reported to trigger a dramatic increase in circulating neutrophils and extracellular DNA traps in 4T1 tumor-bearing mice [12,34,35]

Read more

Summary

Introduction

Cancer is commonly associated with hypercoagulability and a high risk of thrombosis, which make it the second leading cause of mortality in patients with cancer [1]. Anticancer treatments, such as surgery, chemotherapy, angiogenesis inhibitors, and central venous catheters further aggravate cancer-associated thrombosis (CAT) [2]. The treatment of CAT is challenging due to its high risk of recurrence and bleeding caused by anticoagulant therapy [3]. The pathophysiology of thrombosis in cancer is complex, and its regulatory mechanism remains to be elucidated. The mechanisms by which tumors mediate the tissue factor expression in different cell types are largely unknown

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