Network


Latest external collaboration on country level. Dive into details by clicking on the dots.

Hotspot


Dive into the research topics where Jianjun He is active.

Publication


Featured researches published by Jianjun He.


Oncology Reports | 2015

miR-494 suppresses the progression of breast cancer in vitro by targeting CXCR4 through the Wnt/β-catenin signaling pathway

Lingqin Song; Di Liu; Bao-Feng Wang; Jianjun He; Shuqun Zhang; Zhijun Dai; Xiao-Bin Ma; Xi-Jing Wang

Breast cancer is the most common cancer among women with a high mortality worldwide, which is mainly due to tumor invasion and metastasis. Previous studies have reported that microRNA-494 (miR-494) is downregulated in breast cancer cells. The present study investigated the role of miR-494 in the progression of breast cancer and the underlying mechanisms. The levels of miR-494 were analyzed in several breast cancer cell lines by quantitative reverse transcription PCR (qRT-PCR). The miR-494 mRNA levels were significantly lower in the malignant breast cancer cells than the level in the non-malignant normal breast epithelial cells. miR-494 mimic transfection upregulated the expression levels of E-cadherin, yet downregulated N-cadherin, vimentin and α-smooth muscle actin (α-SMA) in the breast cancer cells. As expected, the expression of these markers in breast cancer cells transfected with miR-494 inhibitors exhibited the opposite variation trend. MTT and Transwell assays showed that cell proliferation and invasion were both significantly suppressed by miR-494 mimics, and were significantly promoted by miR-494 inhibitors. The protein expression level of chemokine (C-X-C motif) receptor 4 (CXCR4) in the breast cancer cells was significantly inhibited by miR-494 mimics, and enhanced by miR-494 inhibitors. Yet, the mRNA level of CXCR4 was barely affected by miR-494 mimics or inhibitors. Dual-luciferase assay confirmed that miR-494 directly interacted with the 3-untranslated region of CXCR4 mRNA by dual-luciferase assay. The miR-494 mimics also significantly inhibited the transcription levels of β-catenin, LEF1, CD44 and cyclin-D1, which was similar to the effect of siRNA targeted to CXCR4. In conclusion, miR-494 suppresses the progression of breast cancer through the Wnt/β-catenin signaling pathway, which is mediated by CXCR4.


Scientific Reports | 2015

Vascular Normalization Induced by Sinomenine Hydrochloride Results in Suppressed Mammary Tumor Growth and Metastasis

Huimin Zhang; Yu Ren; Xiaojiang Tang; Ke Wang; Yang Liu; Li Zhang; Xiao Li; Peijun Liu; Changqi Zhao; Jianjun He

Solid tumor vasculature is characterized by structural and functional abnormality and results in a hostile tumor microenvironment that mediates several deleterious aspects of tumor behavior. Sinomenine is an alkaloid extracted from the Chinese medicinal plant, Sinomenium acutum, which has been utilized to treat rheumatism in China for over 2000 years. Though sinomenine has been demonstrated to mediate a wide range of pharmacological actions, few studies have focused on its effect on tumor vasculature. We showed here that intraperitoneally administration of 100u2005mg/kg sinomenine hydrochloride (SH, the hydrochloride chemical form of sinomenine) in two orthotopic mouse breast cancer models for 14 days, delayed mammary tumor growth and decreased metastasis by inducing vascular maturity and enhancing tumor perfusion, while improving chemotherapy and tumor immunity. The effects of SH on tumor vessels were caused in part by its capability to restore the balance between pro-angiogenic factor (bFGF) and anti-angiogenic factor (PF4). However 200u2005mg/kg SH didnt exhibit the similar inhibitory effect on tumor progression due to the immunosuppressive microenvironment caused by excessive vessel pruning, G-CSF upregulation, and GM-CSF downregulation. Altogether, our findings suggest that SH induced vasculature normalization contributes to its anti-tumor and anti-metastasis effect on breast cancer at certain dosage.


Oncotarget | 2017

Sinomenine hydrochloride inhibits breast cancer metastasis by attenuating inflammation-related epithelial-mesenchymal transition and cancer stemness

Xiao Li; Pingping Li; Chao Liu; Yu Ren; Xiaojiang Tang; Ke Wang; Jianjun He

Sinomenine hydrochloride (SH) has been investigated for its anti-tumor growth effect. We have previously reported that SH inhibited breast cancer cell proliferation via MAPKs signaling. However, whether SH could inhibit tumor metastasis has not been fully explored. In this study, we found that SH suppressed the metastasis potential of breast cancer cells. The wound healing and transwell assays showed that SH inhibited the migration and invasion ability of both 4T1 and MDA-MB-231 breast cancer cells. The orthotopic mouse model of 4T1 and the experimental mouse model of MDA-MB-231-luc (MDA-MB-231 cell line expressing firefly luciferase) demonstrated that SH treatment inhibited breast cancer metastasis by inhibiting epithelial–mesenchymal transition (EMT) and cancer stem cell (CSC) properties without obvious hepatotoxicity and renal toxicity. We also found that SH decreased spleen volume and weight in both mouse models, especially in the 4T1 mouse model. IL-6, a strong inflammatory factor causing EMT, was remarkably reduced. Overall, this anti-metastasis effect of SH could be possibly caused by attenuating inflammatory reaction, which led to inhibition of EMT and CSC characteristics of breast cancer cells. This study, together with our previous one, provides more evidence of SH as a potential drug for breast cancer therapy.


Oncology Research | 2017

Tumor Necrosis Factor (TNF)-α-Induced Protein 8-like-2 (TIPE2) Inhibits Proliferation and Tumorigenesis in Breast Cancer Cells.

Ke Wang; Yu Ren; Yang Liu; Jian Zhang; Jianjun He

Tumor necrosis factor-α (TNF-α)-induced protein 8-like-2 (TNFAIP8L2 or TIPE2), a member of the tumor necrosis TNFAIP8 family, was found to be involved in the development and progression of several tumors. However, to date, the role of TIPE2 in breast cancer is still unclear. Thus, the aim of this study is to explore the role of TIPE2 in breast cancer. Our results indicated that TIPE2 expression was significantly decreased in human breast cancer tissue and cell lines. Overexpression of TIPE2 inhibited the proliferation in vitro and tumor xenograft growth in vivo. TIPE2 also inhibited the migration/invasion of breast cancer cells through preventing the epithelial-to-mesenchymal transition (EMT) phenotype. Mechanically, TIPE2 inhibited the expression of β-catenin, cyclin D1, and c-Myc in breast cancer cells. In conclusion, our findings show that TIPE2 may play an important role in breast cancer cell proliferation, invasion, and tumorigenesis in vivo. Therefore, TIPE2 may be a potential molecular target for the treatment of breast cancer.


Oncology Reports | 2012

Establishment of a bioluminescent MDA-MB-231 cell line for human triple-negative breast cancer research

Ke Wang; Simei Xie; Yu Ren; Haibin Xia; Xinwei Zhang; Jianjun He

The aim of this study was to establish a bioluminescent MDA-MB-231 cell line stably expressing luciferase and green fluorescent protein for the generation of a xenografted model of human triple-negative breast cancer (TNBC) in nude mice. Lentivirus vectors carrying eGFP, firefly luc2 and neo fusion genes were used to transduce the MDA-MB-231 human TNBC cells in vitro. After 8 weeks of G418 selection, eGFP and luc2 expression was determined using a fluorescence microscope and a Xenogen IVIS200 bioluminescent imaging system, respectively. The MTT, transwell invasion and wound healing assays were performed to confirm whether cellular proliferation, invasion and migration were altered by lentiviral infection. Cells were orthotopically implanted into female BALB/c nude mice to test the sensitivity and stability of reporter gene expression. Growth of the tumors was monitored with the in vivo imaging system once a week until they were large enough for experiments. The tumor tissues were resected for histology, and cancer cells were harvested for culture. The lentivirus-transduced MDA-MB-231 cells could stably express luc2 and eGFP, and the luciferase activity reached 9689 photons/sec/cell. Meanwhile, no significant difference in biological activities was observed between the lentivirus-transduced MDA-MB-231 cells and parental cells. An orthotopically implanted tumor model of human TNBCs was successfully established in BALB/c nude mice. Lentiviruses may be ideal carriers for luciferase genes due to their highly efficient infectivity and stable transgene expression. The modified MDA-MB-231 cell line stably expressing luciferase could be detected, allowing for immediate and sensitive detection of metastasis sites in nude mice. As the eGFP and luc2 combination are superior to single reporter genes in their ability to mark cells in vivo and in vitro, these cells may provide a visualizable, convenient and sensitive platform for research on the mechanisms of metastasis and the development of new antitumor drugs for human TNBC.


Health and Quality of Life Outcomes | 2016

Health-related quality of life of postmenopausal Chinese women with hormone receptor-positive early breast cancer during treatment with adjuvant aromatase inhibitors: a prospective, multicenter, non-interventional study.

Ayong Cao; Jin Zhang; Xiaoan Liu; Weizhu Wu; Yinhua Liu; Zhimin Fan; Anqin Zhang; Tianning Zhou; Peifen Fu; Shu Wang; Quchang Ouyang; Jinhai Tang; Hongchuan Jiang; Xiaohua Zhang; Da Pang; Jianjun He; Linxiang Shi; Xianming Wang; Yuan Sheng; Dahua Mao; Zhimin Shao

BackgroundEstimating quality of life (QoL) in patients with breast cancer is of importance in assessing treatment outcomes. Adjuvant endocrine therapy is widely used for hormone receptor-positive (HR+) early-stage breast cancer (EBC), and evidence suggests that aromatase inhibitors (AIs) may improve QoL for these patients. This study evaluated QoL in postmenopausal Chinese patients with HR+ EBC taking AIs.MethodsThis was a prospective, multicenter, and observational study that had no intent to intervene in the current treatment of recruited patients. Eligible patients were recruited within 7xa0days of beginning adjuvant treatment with AIs. The Functional Assessment of Cancer Therapy-Breast (FACT-B) scale was used to evaluate the patients’ QoL. Data were collected at baseline and at 6, 12, 18, and 24xa0months.ResultsFrom June 2010 to October 2013, a total of 494 patients with HR+ EBC were recruited from 21 centers. There was a 7.51-point increase in the patients’ mean FACT-B trial outcome index (TOI), from 90.69 at baseline to 98.72 at 24xa0months (Pu2009<u2009.0001). The mean TOI scores at baseline, 6, 12, and 18xa0months were 90.69, 94.36, 97.71, and 96.75, respectively (Pu2009<u2009.0001, for all). The mean (FACT-B) emotional well-being subscale scores at baseline, 6, 12, 18, and 24xa0months were 16.32, 16.55, 17.34 (Pu2009<u2009.0001), 17.47 (Pu2009<u2009.0001), and 17.85 (Pu2009<u2009.0001), respectively, and social well-being scores were 18.61, 19.14 (Pu2009<u2009.04), 19.35 (Pu2009<u2009.008), 18.32, and 18.40, respectively. In the mixed model, baseline TOI, clinical visits, prior chemotherapies, age group, and axillary lymph-node dissection presented statistically significant effects on the change of FACT-B TOI and FACT-B SWB, whereas only baseline TOI, clinical visits, and prior chemotherapies presented statistically significant effects on the change of FACT-B EWB. FACT-B TOI, being the most pertinent and precise indicator of patient-reported QoL, demonstrated significant changes reflecting clinical benefit of adjuvant AIs endocrine therapy in the QoL of HRu2009+u2009EBC patients.ConclusionsThe study demonstrated significant improvements in the long-term QoL of postmenopausal Chinese patients with HR+ EBC at 6, 12, 18, and 24xa0months after starting treatment with AIs. The current study indicates improved long-term QoL with AI adjuvant treatment, which will aid clinicians in optimizing treatment to yield effective healthcare outcomes.Trial registrationClinicaltrials.gov NCT01144572


Oncology Research | 2017

MiR-4262 promotes proliferation and invasion of human breast cancer cells through directly targeting KLF6 and KLF15.

Ke Wang; Yu Ren; Yang Liu; Jian Zhang; Jianjun He

miRNAs have been shown to be involved in breast cancer growth and progression. miR-4262 is a potential tumor promoter in human cancers. In this study, we first investigated the role of miR-4262 in the proliferation and invasion of human breast cancer cells. Our results showed that, compared with the adjacent tissues and MCF-10A normal breast epithelial cells, miR-4262 was markedly increased in the breast cancer tissues and five cell lines, including MDA-MB-231, MDA-MB-468, MDA-MB-435, SKBR3, and MCF-7. Then the miR-4262 mimic or oligo anta-miR-4262 was transfected into MDA-MB-231 and MCF-7 breast cancer cell lines. The results showed that the miR-4262 mimic greatly increased the miR-4262 level and the proliferation and invasion of MDA-MB-231 and MCF-7 cells. In contrast, the anta-miR-4262 had a completely opposite effect on miR-4262 expression, cell proliferation, and cell invasion in MDA-MB-231 and MCF-7 cells. Moreover, bioinformatics and luciferase reporter gene assays confirmed that miR-4262 targeted the mRNA 3-UTR region of KLF6 and KLF15, two characterized tumor suppressor genes. miR-4262 suppressed protein levels of KLF6 and KLF15 in MDA-MB-231 cells, and the suppression could be rescued by the transfection of pcDNA-KLF6 and -KLF15. In conclusion, miR-4262 positively regulates proliferation and invasion of human breast cancer cells via suppression of KLF6 and KLF15.


Apmis | 2016

SOX1 inhibits breast cancer cell growth and invasion through suppressing the Wnt/β-catenin signaling pathway.

Lingqin Song; Di Liu; Jianjun He; Xi-Jing Wang; Zhijun Dai; Yang Zhao; Hua-Feng Kang; Bao-Feng Wang

Abnormal activation of the Wnt/β‐catenin signaling pathway is common in human cancers. Several studies have demonstrated that SRY (sex‐determining region Y)‐box (SOX) family genes serve as either tumor suppressor genes or oncogenes by regulating the Wnt signaling pathway in different cancers. However, the role of SOX1 in breast cancer and the underlying mechanism is still unclear. The aim of this study was to explore the effect and mechanism of SOX1 on the breasted cancer cell growth and invasion. In this study, we established overexpressed SOX1 and investigated its function by in vitro experiments. SOX1 was down‐regulated in breast cancer tissues and cell lines. Overexpression of SOX1 inhibited cell proliferation and invasion in vitro, and it promoted cell apoptosis. Furthermore, SOX1 inhibited the expression of β‐catenin, cyclin D1, and c‐Myc in breast cancer cells. Taken together, these data suggest that SOX1 can function as a tumor suppressor partly by interfering with Wnt/β‐catenin signaling in breast cancer.


Oncotarget | 2017

Cell-free circulating tumor DNA analysis for breast cancer and its clinical utilization as a biomarker

Ru Wang; Xiao Li; Huimin Zhang; Ke Wang; Jianjun He

Circulating tumor DNA (ctDNA) in the blood of cancer patients contains much information on genetic and epigenetic profiles associated with cancer development, progression, and response to therapy. Analysis of ctDNA provides an opportunity for non-invasive sampling of tumor DNA repetitiously and therefore advance precision medicine. Recent development in massively parallel sequencing and digital genomic techniques support the analytical and clinical validity of ctDNA as a promising ‘liquid biopsy’ in human cancer. In this review, we discussed the current status of cell-free ctDNA including ctDNA biology, recently developed techniques for ctDNA detection, breast cancer specific detecting strategies, with a focus on clinical applications of ctDNA-based biomarkers in breast oncology.


Biomedicine & Pharmacotherapy | 2018

Sinomenine reduces growth and metastasis of breast cancer cells and improves the survival of tumor-bearing mice through suppressing the SHh pathway

Lingqin Song; Di Liu; Yang Zhao; Jianjun He; Hua-Feng Kang; Zhijun Dai; Xi-Jing Wang; Shuqun Zhang; Ying Zan; Xinghuan Xue

In this study, the suppressive effect of sinomenine on the activation of SHh and the progression of breast cancer metastasis in vitro and in vivo was investigated. MDA-MB-231 breast cancer cells were treated with sinomenine and/or cyclopamine a proven SHh inhibitor. Sinomenine and cyclopamine both suppressed cell proliferation and migration, but sinomenine had a stronger suppressive effect in MDA-MB-231. In addition, sinomenine could suppress the activation of NF-κB and SHh signaling pathways, but cyclopamine could not suppress the activation of NF-κB. Subsequently, a mouse breast cancer-lung metastasis model was established. Our data on tissue examination and gene detection showed that SHh signaling was markedly activated in the metastatic model mice. The progression of lung metastasis was suppressed when mice were fed sinomenine and/or cyclopamine, while sinomenine had a stronger suppressive effect than cyclopamine in the model mice. In conclusion, sinomenine has a better effect than cyclopamine on the inhibition of breast cancer metastasis to lung in vivo and vitro, and inhibits NF-κB activation and NF-κB-mediated activation of SHh signaling pathway.

Collaboration


Dive into the Jianjun He's collaboration.

Top Co-Authors

Avatar

Yu Ren

Xi'an Jiaotong University

View shared research outputs
Top Co-Authors

Avatar

Ke Wang

Xi'an Jiaotong University

View shared research outputs
Top Co-Authors

Avatar

Yang Liu

Xi'an Jiaotong University

View shared research outputs
Top Co-Authors

Avatar

Di Liu

Xi'an Jiaotong University

View shared research outputs
Top Co-Authors

Avatar

Huimin Zhang

Xi'an Jiaotong University

View shared research outputs
Top Co-Authors

Avatar

Jian Zhang

Xi'an Jiaotong University

View shared research outputs
Top Co-Authors

Avatar

Jianghua Ou

Xinjiang Medical University

View shared research outputs
Top Co-Authors

Avatar

Lingqin Song

Xi'an Jiaotong University

View shared research outputs
Top Co-Authors

Avatar

Xi-Jing Wang

Xi'an Jiaotong University

View shared research outputs
Top Co-Authors

Avatar

Xiao Li

Xi'an Jiaotong University

View shared research outputs
Researchain Logo
Decentralizing Knowledge