Network


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

Hotspot


Dive into the research topics where Lu-Zhe Sun is active.

Publication


Featured researches published by Lu-Zhe Sun.


The Prostate | 2013

Single-cell analysis of circulating tumor cells identifies cumulative expression patterns of EMT-related genes in metastatic prostate cancer

Chun Liang Chen; Devalingam Mahalingam; Pawel A. Osmulski; Rohit R. Jadhav; Chiou Miin Wang; Robin J. Leach; Tien Cheng Chang; Steven Weitman; Addanki P. Kumar; Lu-Zhe Sun; Maria Gaczynska; Ian M. Thompson; Tim H M Huang

Prostate tumors shed circulating tumor cells (CTCs) into the blood stream. Increased evidence shows that CTCs are often present in metastatic prostate cancer and can be alternative sources for disease profiling and prognostication. Here we postulate that CTCs expressing genes related to epithelial–mesenchymal transition (EMT) are strong predictors of metastatic prostate cancer.


PLOS ONE | 2010

Doxorubicin in Combination with a Small TGFβ Inhibitor: A Potential Novel Therapy for Metastatic Breast Cancer in Mouse Models

Abhik Bandyopadhyay; Long Wang; Joseph K. Agyin; Yuping Tang; Shu Lin; I-Tien Yeh; Keya De; Lu-Zhe Sun

Background Recent studies suggested that induction of epithelial-mesenchymal transition (EMT) might confer both metastatic and self-renewal properties to breast tumor cells resulting in drug resistance and tumor recurrence. TGFβ is a potent inducer of EMT and has been shown to promote tumor progression in various breast cancer cell and animal models. Principal Findings We report that chemotherapeutic drug doxorubicin activates TGFβ signaling in human and murine breast cancer cells. Doxorubicin induced EMT, promoted invasion and enhanced generation of cells with stem cell phenotype in murine 4T1 breast cancer cells in vitro, which were significantly inhibited by a TGFβ type I receptor kinase inhibitor (TβRI-KI). We investigated the potential synergistic anti-tumor activity of TβR1-KI in combination with doxorubicin in animal models of metastatic breast cancer. Combination of Doxorubicin and TβRI-KI enhanced the efficacy of doxorubicin in reducing tumor growth and lung metastasis in the 4T1 orthotopic xenograft model in comparison to single treatments. Doxorubicin treatment alone enhanced metastasis to lung in the human breast cancer MDA-MB-231 orthotopic xenograft model and metastasis to bone in the 4T1 orthotopic xenograft model, which was significantly blocked when TβR1-KI was administered in combination with doxorubicin. Conclusions These observations suggest that the adverse activation of TGFβ pathway by chemotherapeutics in the cancer cells together with elevated TGFβ levels in tumor microenvironment may lead to EMT and generation of cancer stem cells resulting in the resistance to the chemotherapy. Our results indicate that the combination treatment of doxorubicin with a TGFβ inhibitor has the potential to reduce the dose and consequently the toxic side-effects of doxorubicin, and improve its efficacy in the inhibition of breast cancer growth and metastasis.


Oncogene | 2014

Androgen receptor and microRNA-21 axis downregulates transforming growth factor beta receptor II (TGFBR2) expression in prostate cancer

Sweta Mishra; Janice Jianhong Deng; Pramod S. Gowda; Manjeet K. Rao; Chun-Lin Lin; Chun Liang Chen; Tim H M Huang; Lu-Zhe Sun

Prostate cancer cells escape growth inhibition from transforming growth factor β (TGFβ) by downregulating TGFβ receptors. However, the mechanism by which cancer cells downregulate TGFβ receptors in prostate is not clear. Here, we showed that coordinated action of miR-21 and androgen receptor (AR) signaling had a critical role in inhibiting TGFβ receptor II (TGFBR2) expression in prostate cancer cells. Our results revealed that miR-21 suppresses TGFBR2 levels by binding to its 3′-UTR and AR signaling further potentiates this effect in both untransformed and transformed human prostate epithelial cells as well as in human prostate cancers. Analysis of primary prostate cancers showed that increased miR-21/AR expression parallel a significantly reduced expression of TGFBR2. Manipulation of androgen signaling or the expression levels of AR or miR-21 negatively altered TGFBR2 expression in untransformed and transformed human prostate epithelial cells, human prostate cancer xenografts and mouse prostate glands. Importantly, we demonstrated that miR-21 and AR regulated each other’s expression resulting in a positive feedback loop. Our results indicated that miR-21/AR mediate its tumor-promoting function by attenuating TGFβ-mediated Smad2/3 activation, cell growth inhibition, cell migration and apoptosis. Together, these results suggest that the AR and miR-21 axis exerts its oncogenic effects in prostate tumors by downregulating TGFBR2, hence inhibiting the tumor-suppressive activity of TGFβ pathway. Targeting miR-21 alone or in combination with AR may restore the tumor inhibitory activity of TGFβ in prostate cancer.


Cancer Research | 2010

Extranuclear Functions of ER Impact Invasive Migration and Metastasis by Breast Cancer Cells

Dimple Chakravarty; Sujit S. Nair; Bindu Santhamma; Binoj C. Nair; Long Wang; Abhik Bandyopadhyay; Joseph K. Agyin; Darrell W. Brann; Lu-Zhe Sun; I-Tien Yeh; Francis Y. Lee; Rajeshwar Rao Tekmal; Rakesh Kumar; Ratna K. Vadlamudi

The molecular basis of breast cancer progression to metastasis and the role of estrogen receptor (ER) signaling in this process remain poorly understood. Emerging evidence suggests that ER participates in extranuclear signaling in addition to genomic functions. Recent studies identified proline-, glutamic acid-, and leucine-rich protein-1 (PELP1) as one of the components of ER signalosome in the cytoplasm. PELP1 expression is deregulated in metastatic breast tumors. We examined the mechanism and significance of ER-PELP1-mediated extranuclear signals in the cytoskeletal remodeling and metastasis. Using estrogen dendrimer conjugate (EDC) that uniquely activate ER extranuclear signaling and by using model cells that stably express PELP1 short hairpin RNA (shRNA), we show that PELP1 is required for optimal activation of ER extranuclear actions. Using a yeast two-hybrid screen, we identified integrin-linked kinase 1 (ILK1) as a novel PELP1-binding protein. Activation of extranuclear signaling by EDC uniquely enhanced E2-mediated ruffles and filopodia-like structures. Using dominant-negative and dominant-active reagents, we found that estrogen-mediated extranuclear signaling promotes cytoskeleton reorganization through the ER-Src-PELP1-phosphoinositide 3-kinase-ILK1 pathway. Using in vitro Boyden chamber assays and in vivo xenograft assays, we found that ER extranuclear actions contribute to cell migration. Collectively, our results suggest that ER extranuclear actions play a role in cell motility/metastasis, establishing for the first time that endogenous PELP1 serves as a critical component of ER extranuclear actions leading to cell motility/invasion and that the ER-Src-PELP1-ILK1 pathway represents a novel therapeutic target for preventing the emergence of ER-positive metastasis.


The EMBO Journal | 2011

TGF-β signalling is mediated by two autonomously functioning TβRI:TβRII pairs

Tao Huang; Laurent David; Valentín Mendoza; Yong Yang; Maria M. Villarreal; Keya De; Lu-Zhe Sun; Xiaohong Fang; Fernando López-Casillas; Jeffrey L. Wrana; Andrew P. Hinck

Transforming growth factor (TGF)‐βs are dimeric polypeptides that have vital roles in regulating cell growth and differentiation. They signal by assembling a receptor heterotetramer composed of two TβRI:TβRII heterodimers. To investigate whether the two heterodimers bind and signal autonomously, one of the TGF‐β protomers was substituted to block receptor binding. The substituted dimer, TGF‐β3 WD, bound the TβRII extracellular domain and recruited the TβRI with affinities indistinguishable from TGF‐β3, but with one‐half the stoichiometry. TGF‐β3 WD was further shown to retain one‐quarter to one‐half the signalling activity of TGF‐β3 in three established assays for TGF‐β function. Single‐molecule fluorescence imaging with GFP‐tagged receptors demonstrated a measurable increase in the proportion of TβRI and TβRII dimers upon treatment with TGF‐β3, but not with TGF‐β3 WD. These results provide evidence that the two TβRI:TβRII heterodimers bind and signal in an autonomous manner. They further underscore how the TGF‐βs diverged from the bone morphogenetic proteins, the ancestral ligands of the TGF‐β superfamily that signal through a RI:RII:RII heterotrimer.


Oncogene | 2014

Significance of PELP1/HDAC2/miR-200 regulatory network in EMT and metastasis of breast cancer

Sudipa Saha Roy; Vk Gonugunta; Abhik Bandyopadhyay; Manjeet K. Rao; Gregory J. Goodall; Lu-Zhe Sun; Rajeshwar Rao Tekmal; Ratna K. Vadlamudi

Tumor metastasis is the leading cause of death among breast cancer patients. PELP1 (proline, glutamic acid and leucine rich protein 1) is a nuclear receptor coregulator that is upregulated during breast cancer progression to metastasis and is an independent prognostic predictor of shorter survival of breast cancer patients. Here, we show that PELP1 modulates expression of metastasis-influencing microRNAs (miRs) to promote cancer metastasis. Whole genome miR array analysis using PELP1-overexpressing and PELP1-underexpressing model cells revealed that miR-200 and miR-141 levels inversely correlated with PELP1 expression. Consistent with this, PELP1 knockdown resulted in lower expression of miR-200a target genes ZEB1 and ZEB2. PELP1 knockdown significantly reduced tumor growth and metastasis compared with parental cells in an orthotopic xenograft tumor model. Furthermore, re-introduction of miR-200a and miR-141 mimetics into PELP1-overexpressing cells reversed PELP1 target gene expression, decreased PELP1-driven migration/invasion in vitro and significantly reduced in vivo metastatic potential in a preclinical model of experimental metastasis. Our results demonstrated that PELP1 binds to miR-200a and miR-141 promoters and regulates their expression by recruiting chromatin modifier histone deacetylase 2 (HDAC2) as revealed by chromatin immunoprecipitation, small interfering RNA and HDAC inhibitor assays. Taken together, our results suggest that PELP1 regulates tumor metastasis by controlling the expression and functions of the tumor metastasis suppressors miR-200a and miR-141.


Oncogene | 2015

Differential impact of adenosine nucleotides released by osteocytes on breast cancer growth and bone metastasis

Jade Z. Zhou; Manuel A. Riquelme; Xiaoli Gao; Lesley G. Ellies; Lu-Zhe Sun; Jean X. Jiang

Extracellular ATP has been shown to either inhibit or promote cancer growth and migration; however, the mechanism underlying this discrepancy remained elusive. Here we demonstrate the divergent roles of ATP and adenosine released by bone osteocytes on breast cancers. We showed that conditioned media (CM) collected from osteocytes treated with alendronate (AD), a bisphosphonate drug, inhibited the migration of human breast cancer MDA-MB-231 cells. Removal of the extracellular ATP by apyrase in CM abolished this effect, suggesting the involvement of ATP. ATP exerted its inhibitory effect through the activation of purinergic P2X receptor signaling in breast cancer cells evidenced by the attenuation of the inhibition by an antagonist, oxidized ATP, as well as knocking down P2X7 with small interfering RNA (siRNA), and the inhibition of migration by an agonist, BzATP. Intriguingly, ATP had a biphasic effect on breast cancer cells—lower dosage inhibited but higher dosage promoted its migration. The stimulatory effect on migration was blocked by an adenosine receptor antagonist, MRS1754, ARL67156, an ecto-ATPase inhibitor, and A2A receptor siRNA, suggesting that in contrast to ATP, adenosine, a metabolic product of ATP, promoted migration of breast cancer cells. Consistently, non-hydrolyzable ATP, ATPγS, only inhibited but did not promote cancer cell migration. ATP also had a similar inhibitory effect on the Py8119 mouse mammary carcinoma cells; however, adenosine had no effect owing to the absence of the A2A receptor. Consistently, ATPγS inhibited, whereas adenosine promoted anchorage-independent growth of MDA-MB-231 cells. Our in vivo xenograft study showed a significant delay of tumor growth with the treatment of ATPγS. Moreover, the extent of bone metastasis in a mouse intratibial model was significantly reduced with the treatment of ATPγS. Together, our results suggest the distinct roles of ATP and adenosine released by osteocytes and the activation of corresponding receptors P2X7 and A2A signaling on breast cancer cell growth, migration and bone metastasis.


PLOS ONE | 2011

Reduced expression of fumarate hydratase in clear cell renal cancer mediates HIF-2α accumulation and promotes migration and invasion.

Sunil Sudarshan; Karthigayan Shanmugasundaram; Susan L. Naylor; Shu Lin; Carolina B. Livi; Christine F. O'Neill; Dipen J. Parekh; I-Tien Yeh; Lu-Zhe Sun; Karen Block

Germline mutations of FH, the gene that encodes for the tricarboxylic acid TCA (TCA) cycle enzyme fumarate hydratase, are associated with an inherited form of cancer referred to as Hereditary Leiomyomatosis and Renal Cell Cancer (HLRCC). Individuals with HLRCC are predisposed to the development of highly malignant and lethal renal cell carcinoma (RCC). The mechanisms of tumorigenesis proposed have largely focused on the biochemical consequences of loss of FH enzymatic activity. While loss of the tumor suppressor gene von Hippel Lindau (VHL) is thought to be an initiating event for the majority of RCCs, a role for FH in sporadic renal cancer has not been explored. Here we report that FH mRNA and protein expression are reduced in clear cell renal cancer, the most common histologic variant of kidney cancer. Moreover, we demonstrate that reduced FH leads to the accumulation of hypoxia inducible factor- 2α (HIF-2α), a transcription factor known to promote renal carcinogenesis. Finally, we demonstrate that overexpression of FH in renal cancer cells inhibits cellular migration and invasion. These data provide novel insights into the tumor suppressor functions of FH in sporadic kidney cancer.


Molecular Cancer Research | 2012

Significance of PELP1 in ER-Negative Breast Cancer Metastasis

Sudipa Saha Roy; Dimple Chakravarty; Valerie Cortez; Keya De Mukhopadhyay; Abhik Bandyopadhyay; Jung Mo Ahn; Ganesh V. Raj; Rajeshwar Rao Tekmal; Lu-Zhe Sun; Ratna K. Vadlamudi

Breast cancer metastasis is a major clinical problem. The molecular basis of breast cancer progression to metastasis remains poorly understood. PELP1 is an estrogen receptor (ER) coregulator that has been implicated as a proto-oncogene whose expression is deregulated in metastatic breast tumors and whose expression is retained in ER-negative tumors. We examined the mechanism and significance of PELP1-mediated signaling in ER-negative breast cancer progression using two ER-negative model cells (MDA-MB-231 and 4T1 cells) that stably express PELP1-shRNA. These model cells had reduced PELP1 expression (75% of endogenous levels) and exhibited less propensity to proliferate in growth assays in vitro. PELP1 downregulation substantially affected migration of ER-negative cells in Boyden chamber and invasion assays. Using mechanistic studies, we found that PELP1 modulated expression of several genes involved in the epithelial mesenchymal transition (EMT), including MMPs, SNAIL, TWIST, and ZEB. In addition, PELP1 knockdown reduced the in vivo metastatic potential of ER-negative breast cancer cells and significantly reduced lung metastatic nodules in a xenograft assay. These results implicate PELP1 as having a role in ER-negative breast cancer metastasis, reveal novel mechanism of coregulator regulation of metastasis via promoting cell motility/EMT by modulating expression of genes, and suggest PELP1 may be a potential therapeutic target for metastatic ER-negative breast cancer. Mol Cancer Res; 10(1); 25–33. ©2011 AACR.


Molecular Biology of the Cell | 2012

Attenuation of TGF-β signaling suppresses premature senescence in a p21-dependent manner and promotes oncogenic Ras-mediated metastatic transformation in human mammary epithelial cells

Shu Lin; Junhua Yang; Abdel G. Elkahloun; Abhik Bandyopadhyay; Long Wang; John E. Cornell; I-Tien Yeh; Joseph K. Agyin; Gail E. Tomlinson; Lu-Zhe Sun

A series of isogenic, basal-like human mammary epithelial cells (HMECs) with altered TGF-β sensitivity and different malignancy is used to elucidate molecular mechanisms that evade oncogenic Ras-induced growth arrest and promote transformation. Attenuation of TGF-β signaling is found to cause metastatic progression of Ras-transformed HMECs.

Collaboration


Dive into the Lu-Zhe Sun's collaboration.

Top Co-Authors

Avatar

Abhik Bandyopadhyay

University of Texas Health Science Center at San Antonio

View shared research outputs
Top Co-Authors

Avatar

Junhua Yang

University of Texas Health Science Center at San Antonio

View shared research outputs
Top Co-Authors

Avatar

Xiang Gu

University of Texas Health Science Center at San Antonio

View shared research outputs
Top Co-Authors

Avatar

Shu Lin

University of Texas Health Science Center at San Antonio

View shared research outputs
Top Co-Authors

Avatar

Sweta Mishra

University of Texas Health Science Center at San Antonio

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Hakim Bouamar

University of Texas Health Science Center at San Antonio

View shared research outputs
Top Co-Authors

Avatar

Long Wang

University of Texas Health Science Center at San Antonio

View shared research outputs
Top Co-Authors

Avatar

Zhao Liu

Nanjing Medical University

View shared research outputs
Researchain Logo
Decentralizing Knowledge