Network


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

Hotspot


Dive into the research topics where Xiaona Huang is active.

Publication


Featured researches published by Xiaona Huang.


Oncotarget | 2016

Cancer therapies activate RIG-I-like receptor pathway through endogenous non-coding RNAs

Diana Rose E. Ranoa; A. Parekh; Sean P. Pitroda; Xiaona Huang; Thomas E. Darga; Antony C. Wong; Lei Huang; Jorge Andrade; Jonathan P. Staley; Takashi Satoh; Shizuo Akira; Ralph R. Weichselbaum; Nikolai N. Khodarev

Emerging evidence indicates that ionizing radiation (IR) and chemotherapy activate Type I interferon (IFN) signaling in tumor and host cells. However, the mechanism of induction is poorly understood. We identified a novel radioprotective role for the DEXH box RNA helicase LGP2 (DHX58) through its suppression of IR-induced cytotoxic IFN-beta [1]. LGP2 inhibits activation of the RIG-I-like receptor (RLR) pathway upon binding of viral RNA to the cytoplasmic sensors RIG-I (DDX58) and MDA5 (IFIH1) and subsequent IFN signaling via the mitochondrial adaptor protein MAVS (IPS1). Here we show that MAVS is necessary for IFN-beta induction and interferon-stimulated gene expression in the response to IR. Suppression of MAVS conferred radioresistance in normal and cancer cells. Germline deletion of RIG-I, but not MDA5, protected mice from death following total body irradiation, while deletion of LGP2 accelerated the death of irradiated animals. In human tumors depletion of RIG-I conferred resistance to IR and different classes of chemotherapy drugs. Mechanistically, IR stimulated the binding of cytoplasmic RIG-I with small endogenous non-coding RNAs (sncRNAs), which triggered IFN-beta activity. We demonstrate that the small nuclear RNAs U1 and U2 translocate to the cytoplasm after IR treatment, thus stimulating the formation of RIG-I: RNA complexes and initiating downstream signaling events. Taken together, these findings suggest that the physiologic responses to radio-/chemo-therapy converge on an antiviral program in recruitment of the RLR pathway by a sncRNA-dependent activation of RIG-I which commences cytotoxic IFN signaling. Importantly, activation of interferon genes by radiation or chemotherapy is associated with a favorable outcome in patients undergoing treatment for cancer. To our knowledge, this is the first demonstration of a cell-intrinsic response to clinically relevant genotoxic treatments mediated by an RNA-dependent mechanism.


Proceedings of the National Academy of Sciences of the United States of America | 2014

RIG-I–like receptor LGP2 protects tumor cells from ionizing radiation

Ryan C. Widau; A. Parekh; M.C. Ranck; Daniel W. Golden; Kiran A. Kumar; Ravi F. Sood; Sean P. Pitroda; Zhengkai Liao; Xiaona Huang; Thomas E. Darga; David Xu; Lei Huang; Jorge Andrade; Bernard Roizman; Ralph R. Weichselbaum; Nikolai N. Khodarev

Significance An undesirable outcome of radiotherapy (ionizing radiation, IR) of cancer is the emergence of radioresistant cells. We report that Laboratory of Genetics and Physiology 2 (LGP2), a resident RIG-I (retinoic acid inducible gene I)–like receptor protein, can induce radioresistance. IR induces interferon and stimulates accumulation of LGP2. In turn, LGP2 shuts off the synthesis of interferon and blocks its cytotoxic effects. Ectopic expression of LGP2 enhances resistance to IR, whereas depletion enhances cytotoxic effects of IR. Here we show that LGP2 is associated with radioresistance in numerous diverse cancer cell lines. Examination of available databases links expression of LGP2 with poor prognosis in cancer patients. An siRNA screen targeting 89 IFN stimulated genes in 14 different cancer cell lines pointed to the RIG-I (retinoic acid inducible gene I)–like receptor Laboratory of Genetics and Physiology 2 (LGP2) as playing a key role in conferring tumor cell survival following cytotoxic stress induced by ionizing radiation (IR). Studies on the role of LGP2 revealed the following: (i) Depletion of LGP2 in three cancer cell lines resulted in a significant increase in cell death following IR, (ii) ectopic expression of LGP2 in cells increased resistance to IR, and (iii) IR enhanced LGP2 expression in three cell lines tested. Studies designed to define the mechanism by which LGP2 acts point to its role in regulation of IFNβ. Specifically (i) suppression of LGP2 leads to enhanced IFNβ, (ii) cytotoxic effects following IR correlated with expression of IFNβ inasmuch as inhibition of IFNβ by neutralizing antibody conferred resistance to cell death, and (iii) mouse embryonic fibroblasts from IFN receptor 1 knockout mice are radioresistant compared with wild-type mouse embryonic fibroblasts. The role of LGP2 in cancer may be inferred from cumulative data showing elevated levels of LGP2 in cancer cells are associated with more adverse clinical outcomes. Our results indicate that cytotoxic stress exemplified by IR induces IFNβ and enhances the expression of LGP2. Enhanced expression of LGP2 suppresses the IFN stimulated genes associated with cytotoxic stress by turning off the expression of IFNβ.


British Journal of Cancer | 2015

Oncogenic CXCL10 signalling drives metastasis development and poor clinical outcome

Sean C. Wightman; Abhineet Uppal; Sean P. Pitroda; Sabha Ganai; Byron Burnette; Melinda E. Stack; Go Oshima; Sajid A. Khan; Xiaona Huang; Mitchell C. Posner; Ralph R. Weichselbaum; Nikolai N. Khodarev

Background:The CXCL10/CXCR3 signalling mediates paracrine interactions between tumour and stromal cells that govern leukocyte trafficking and angiogenesis. Emerging data implicate noncanonical CXCL10/CXCR3 signalling in tumourigenesis and metastasis. However, little is known regarding the role for autocrine CXCL10/CXCR3 signalling in regulating the metastatic potential of individual tumour clones.Methods:We performed transcriptomic and cytokine profiling to characterise the functions of CXCL10 and CXCR3 in tumour cells with different metastatic abilities. We modulated the expression of the CXCL10/CXCR3 pathway using shRNA-mediated silencing in both in vitro and in vivo models of B16F1 melanoma. In addition, we examined the expression of CXCL10 and CXCR3 and their associations with clinical outcomes in clinical data sets derived from over 670 patients with melanoma and colon and renal cell carcinomas.Results:We identified a critical role for autocrine CXCL10/CXCR3 signalling in promoting tumour cell growth, motility and metastasis. Analysis of publicly available clinical data sets demonstrated that coexpression of CXCL10 and CXCR3 predicted an increased metastatic potential and was associated with early metastatic disease progression and poor overall survival.Conclusion:These findings support the potential for CXCL10/CXCR3 coexpression as a predictor of metastatic recurrence and point towards a role for targeting of this oncogenic axis in the treatment of metastatic disease.


Nature Communications | 2017

Host STING-dependent MDSC mobilization drives extrinsic radiation resistance

Hua Liang; Liufu Deng; Yuzhu Hou; Xiangjiao Meng; Xiaona Huang; Enyu Rao; Wenxin Zheng; Helena J. Mauceri; Matthias Mack; Meng Xu; Yang-Xin Fu; Ralph R. Weichselbaum

Radiotherapy induces and promotes innate and adaptive immunity in which host STING plays an important role. However, radioresistance in irradiated tumors can also develop, resulting in relapse. Here we report a mechanism by which extrinsic resistance develops after local ablative radiation that relies on the immunosuppressive action of STING. The STING/type I interferon pathway enhances suppressive inflammation in tumors by recruiting myeloid cells in part via the CCR2 pathway. Germ-line knockouts of CCR2 or treatment with an anti-CCR2 antibody results in blockade of radiation-induced MDSC infiltration. Treatment with anti-CCR2 antibody alleviates immunosuppression following activation of the STING pathway, enhancing the anti-tumor effects of STING agonists and radiotherapy. We propose that radiation-induced STING activation is immunosuppressive due to (monocytic) M-MDSC infiltration, which results in tumor radioresistance. Furthermore, the immunosuppressive effects of radiotherapy and STING agonists can be abrogated in humans by a translational strategy involving anti-CCR2 antibody treatment to improve radiotherapy.Tumors often develop resistance to radiotherapy. Here the authors show that irradiation leads to a CCR2-dependent infiltration by myeloid derived suppressor cells that promote radio-resistance through inhibition of adaptive immune responses and that the use of CCR2 antibodies in mice reduces such resistance.


Scientific Reports | 2016

Basal Tumor Cell Isolation and Patient-Derived Xenograft Engraftment Identify High-Risk Clinical Bladder Cancers

Kinga B. Skowron; Sean P. Pitroda; Jukes P. Namm; O. Balogun; M. A. Beckett; M. L. Zenner; O. Fayanju; Xiaona Huang; Christian Fernandez; Wenxin Zheng; G. Qiao; Robert K. Chin; Stephen J. Kron; Nikolai N. Khodarev; Mitchell C. Posner; Gary D. Steinberg; Ralph R. Weichselbaum

Strategies to identify tumors at highest risk for treatment failure are currently under investigation for patients with bladder cancer. We demonstrate that flow cytometric detection of poorly differentiated basal tumor cells (BTCs), as defined by the co-expression of CD90, CD44 and CD49f, directly from patients with early stage tumors (T1-T2 and N0) and patient-derived xenograft (PDX) engraftment in locally advanced tumors (T3-T4 or N+) predict poor prognosis in patients with bladder cancer. Comparative transcriptomic analysis of bladder tumor cells isolated from PDXs indicates unique patterns of gene expression during bladder tumor cell differentiation. We found cell division cycle 25C (CDC25C) overexpression in poorly differentiated BTCs and determined that CDC25C expression predicts adverse survival independent of standard clinical and pathologic features in bladder cancer patients. Taken together, our findings support the utility of BTCs and bladder cancer PDX models in the discovery of novel molecular targets and predictive biomarkers for personalizing oncology care for patients.


Scientific Reports | 2015

Imaging of tumor clones with differential liver colonization

Go Oshima; Sean C. Wightman; Abhineet Uppal; Melinda E. Stack; Sean P. Pitroda; Jonathan J. Oskvarek; Xiaona Huang; Mitchell C. Posner; Samuel Hellman; Ralph R. Weichselbaum; Nikolai N. Khodarev

We present a model of hepatic colorectal metastases which represents monoclonal cell lines double-labeled by luciferase and tdTomato. These cells form liver metastasis in varying numbers and patterns similar to those observed in patients. Using in vivo and ex vivo luminescent and fluorescent imaging we determine the growth kinetics and clonogenic frequency of tumor cells colonizing liver. Molecular profiling detected stable expressional differences between clones consistent with their phenotypes. The data indicate that clinically relevant phenotypes of liver metastases can be modeled in vivo.


Molecular Cancer Therapeutics | 2018

JAK2 Inhibitor SAR302503 Abrogates PD-L1 Expression and Targets Therapy-Resistant Non–small Cell Lung Cancers

Sean P. Pitroda; Melinda E. Stack; Gene-Fu Liu; Sui-Sui Song; Lucy F. Chen; Hua Liang; A. Parekh; Xiaona Huang; Paul Roach; Mitchell C. Posner; Ralph R. Weichselbaum; Nikolai N. Khodarev

Lung cancer is the leading cause of cancer-related deaths worldwide. Approximately 85% of all lung cancers are non–small cell histology [non–small cell lung cancer (NSCLC)]. Modern treatment strategies for NSCLC target driver oncogenes and immune checkpoints. However, less than 15% of patients survive beyond 5 years. Here, we investigated the effects of SAR302503 (SAR), a selective JAK2 inhibitor, on NSCLC cell lines and tumors. We show that SAR is cytotoxic to NSCLC cells, which exhibit resistance to genotoxic therapies, such as ionizing radiation, cisplatin, and etoposide. We demonstrate that constitutive IFN-stimulated gene expression, including an IFN-related DNA damage resistance signature, predicts for sensitivity to SAR. Importantly, tumor cell–intrinsic expression of PD-L1 is IFN-inducible and abrogated by SAR. Taken together, these findings suggest potential dual roles for JAK2 inhibitors, both as a novel monotherapy in NSCLCs resistant to genotoxic therapies, and in tandem with immune checkpoint inhibition. Mol Cancer Ther; 17(4); 732–9. ©2018 AACR.


Immunity | 2018

Non-canonical NF-κB Antagonizes STING Sensor-Mediated DNA Sensing in Radiotherapy

Yuzhu Hou; Hua Liang; Enyu Rao; Wenxin Zheng; Xiaona Huang; Liufu Deng; Yuan Zhang; Xinshuang Yu; Meng Xu; Helena J. Mauceri; Ainhoa Arina; Ralph R. Weichselbaum; Yang-Xin Fu

SUMMARY The NF‐&kgr;B pathway plays a crucial role in supporting tumor initiation, progression, and radioresistance of tumor cells. However, the role of the NF‐&kgr;B pathway in radiation‐induced anti‐tumor host immunity remains unclear. Here we demonstrated that inhibiting the canonical NF‐&kgr;B pathway dampened the therapeutic effect of ionizing radiation (IR), whereas non‐canonical NF‐&kgr;B deficiency promoted IR‐induced anti‐tumor immunity. Mechanistic studies revealed that non‐canonical NF‐&kgr;B signaling in dendritic cells (DCs) was activated by the STING sensor‐dependent DNA‐sensing pathway. By suppressing recruitment of the transcription factor RelA onto the Ifnb promoter, activation of the non‐canonical NF‐&kgr;B pathway resulted in decreased type I IFN expression. Administration of a specific inhibitor of the non‐canonical NF‐&kgr;B pathway enhanced the anti‐tumor effect of IR in murine models. These findings reveal the potentially interactive roles for canonical and non‐canonical NF‐&kgr;B pathways in IR‐induced STING‐IFN production and provide an alternative strategy to improve cancer radiotherapy. Graphical Abstract Figure. No caption available. HIGHLIGHTSCanonical NF‐&kgr;B pathway is required for radiation‐induced anti‐tumor immunityNon‐canonical NF‐&kgr;B deficiency promotes anti‐tumor immunity after radiotherapyNon‐canonical NF‐&kgr;B pathway inhibits radiation‐induced STING‐mediated type I IFNsInhibiting non‐canonical NF‐&kgr;B pathway potentiates the therapeutic effect of radiation &NA; It is known that the NF‐&kgr;B pathway plays a crucial role in supporting tumor initiation, progression, and the radioresistance of tumor cells. Hou et al. demonstrate that the deficiency of non‐canonical NF‐&kgr;B, but not canonical NF‐&kgr;B, promotes radiation‐induced anti‐tumor immunity by regulating the STING‐mediated type I IFN expression.


Immunity | 2014

STING-dependent cytosolic DNA sensing promotes radiation-induced type I interferon-dependent antitumor immunity in immunogenic tumors

Liufu Deng; Hua Liang; Meng Xu; Xuanming Yang; Byron Burnette; Ainhoa Arina; Xiao Dong Li; Helena J. Mauceri; Michael A. Beckett; Thomas E. Darga; Xiaona Huang; Thomas F. Gajewski; Zhijian J. Chen; Yang-Xin Fu; Ralph R. Weichselbaum


Oncotarget | 2015

14q32-encoded microRNAs mediate an oligometastatic phenotype

Abhineet Uppal; Sean C. Wightman; Stephen Mallon; Go Oshima; Sean P. Pitroda; Qingbei Zhang; Xiaona Huang; Thomas E. Darga; Lei Huang; Jorge Andrade; Huiping Liu; Mark K. Ferguson; Geoffrey L. Greene; Mitchell C. Posner; Samuel Hellman; Nikolai N. Khodarev; Ralph R. Weichselbaum

Collaboration


Dive into the Xiaona Huang's collaboration.

Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Go Oshima

University of Chicago

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

A. Parekh

University of Chicago

View shared research outputs
Top Co-Authors

Avatar

Hua Liang

University of Chicago

View shared research outputs
Researchain Logo
Decentralizing Knowledge