Network


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

Hotspot


Dive into the research topics where Zhengyu Jiang is active.

Publication


Featured researches published by Zhengyu Jiang.


Cell Stem Cell | 2016

Dclk1 Defines Quiescent Pancreatic Progenitors that Promote Injury-Induced Regeneration and Tumorigenesis

C. Benedikt Westphalen; Yoshihiro Takemoto; Takayuki Tanaka; Marina Macchini; Zhengyu Jiang; Bernhard W. Renz; Xiaowei Chen; Steffen Ormanns; Karan Nagar; Yagnesh Tailor; Randal May; Youngjin Cho; Samuel Asfaha; Daniel L. Worthley; Yoku Hayakawa; Aleksandra M. Urbanska; Michael Quante; Maximilian Reichert; Joshua Broyde; Prem S. Subramaniam; Helen Remotti; Gloria H. Su; Anil K. Rustgi; Richard A. Friedman; Barry Honig; Courtney W. Houchen; Kenneth P. Olive; Timothy C. Wang

The existence of adult pancreatic progenitor cells has been debated. While some favor the concept of facultative progenitors involved in homeostasis and repair, neither a location nor markers for such cells have been defined. Using genetic lineage tracing, we show that Doublecortin-like kinase-1 (Dclk1) labels a rare population of long-lived, quiescent pancreatic cells. In vitro, Dclk1+ cells proliferate readily and sustain pancreatic organoid growth. In vivo, Dclk1+ cells are necessary for pancreatic regeneration following injury and chronic inflammation. Accordingly, their loss has detrimental effects after cerulein-induced pancreatitis. Expression of mutant Kras in Dclk1+ cells does not affect their quiescence or longevity. However, experimental pancreatitis converts Kras mutant Dclk1+ cells into potent cancer-initiating cells. As a potential effector of Kras, Dclk1 contributes functionally to the pathogenesis of pancreatic cancer. Taken together, these observations indicate that Dclk1 marks quiescent pancreatic progenitors that are candidates for the origin of pancreatic cancer.


Cancer Cell | 2017

Nerve Growth Factor Promotes Gastric Tumorigenesis through Aberrant Cholinergic Signaling

Yoku Hayakawa; Kosuke Sakitani; Mitsuru Konishi; Samuel Asfaha; Ryota Niikura; Hiroyuki Tomita; Bernhard W. Renz; Yagnesh Tailor; M Macchini; Moritz Middelhoff; Zhengyu Jiang; Takayuki Tanaka; Zinaida A. Dubeykovskaya; Woosook Kim; Xiaowei Chen; Aleksandra M. Urbanska; Karan Nagar; Christoph B. Westphalen; Michael Quante; Chyuan-Sheng Lin; Michael D. Gershon; Akira Hara; Chun-Mei Zhao; Duan Chen; Daniel L. Worthley; Kazuhiko Koike; Timothy C. Wang

Within the gastrointestinal stem cell niche, nerves help to regulate both normal and neoplastic stem cell dynamics. Here, we reveal the mechanisms underlying the cancer-nerve partnership. We find that Dclk1+ tuft cells and nerves are the main sources of acetylcholine (ACh) within the gastric mucosa. Cholinergic stimulation of the gastric epithelium induced nerve growth factor (NGF) expression, and in turn NGF overexpression within gastric epithelium expanded enteric nerves and promoted carcinogenesis. Ablation of Dclk1+ cells or blockade of NGF/Trk signaling inhibited epithelial proliferation and tumorigenesis in an ACh muscarinic receptor-3 (M3R)-dependent manner, in part through suppression of yes-associated protein (YAP) function. This feedforward ACh-NGF axis activates the gastric cancer niche and offers a compelling target for tumor treatment and prevention.


Oncotarget | 2015

IL-17 producing mast cells promote the expansion of myeloid-derived suppressor cells in a mouse allergy model of colorectal cancer

Xiaowei Chen; Michael Churchill; Karan Nagar; Yagnesh Tailor; Timothy H. Chu; Brittany S. Rush; Zhengyu Jiang; Edwin Wang; Bernhard W. Renz; Hongshan Wang; Ming Chiu Fung; Daniel L. Worthley; Siddhartha Mukherjee; Timothy C. Wang

Food allergy can influence the development of colorectal cancer, although the underlying mechanisms are unclear. While mast cells (MC) store and secrete histamine, immature myeloid cells (IMC) are the major site of histidine decarboxylase (HDC) expression, the enzyme responsible for histamine production. From our earlier work, we hypothesized that histamine is central to the association between allergy and colorectal carcinogenesis through its influence on the MC-MDSC axis. Here, we show that in wild type (WT) mice, ovalbumin (OVA) immunization elicits a typical TH2 response. In contrast, in HDC−/− mice, the response to OVA allergy is skewed towards infiltration by IL-17 expressing MCs. This response is inhibited by histamine treatment. The HDC−/− allergic IL-17-expressing MCs promote MDSC proliferation and upregulation of Cox-2 and Arg-1. OVA allergy in HDC−/− mice increases the growth of colon tumor cells in both the MC38 tumor cell implantation model and the AOM/DSS carcinogenesis model. Taken together, our results show that histamine represses IL-17-expressing MCs and their subsequent activation of MDSCs, attenuating the risk of colorectal cancer in the setting of food allergy. Targeting the MC-MDSC axis may be useful for cancer prevention and treatment in patients, particularly in those with food allergy.


Cancer Cell | 2018

β2 Adrenergic-Neurotrophin Feedforward Loop Promotes Pancreatic Cancer

Bernhard W. Renz; Ryota Takahashi; Takayuki Tanaka; M Macchini; Yoku Hayakawa; Zahra Dantes; H. Carlo Maurer; Xiaowei Chen; Zhengyu Jiang; C. Benedikt Westphalen; Matthias Ilmer; Giovanni Valenti; Sarajo Mohanta; Andreas J.R. Habenicht; Moritz Middelhoff; Timothy H. Chu; Karan Nagar; Yagnesh Tailor; Riccardo Casadei; Mariacristina Di Marco; Axel Kleespies; Richard A. Friedman; Helen Remotti; Maximilian Reichert; Daniel L. Worthley; Jens Neumann; Jens Werner; Alina C. Iuga; Kenneth P. Olive; Timothy C. Wang

Catecholamines stimulate epithelial proliferation, but the role of sympathetic nerve signaling in pancreatic ductal adenocarcinoma (PDAC) is poorly understood. Catecholamines promoted ADRB2-dependent PDAC development, nerve growth factor (NGF) secretion, and pancreatic nerve density. Pancreatic Ngf overexpression accelerated tumor development in LSL-Kras+/G12D;Pdx1-Cre (KC) mice. ADRB2 blockade together with gemcitabine reduced NGF expression and nerve density, and increased survival of LSL-Kras+/G12D;LSL-Trp53+/R172H;Pdx1-Cre (KPC) mice. Therapy with a Trk inhibitor together with gemcitabine also increased survival of KPC mice. Analysis of PDAC patient cohorts revealed a correlation between brain-derived neurotrophic factor (BDNF) expression, nerve density, and increased survival of patients on nonselective β-blockers. These findings suggest that catecholamines drive a feedforward loop, whereby upregulation of neurotrophins increases sympathetic innervation and local norepinephrine accumulation.


Gastroenterology | 2018

Immune Cell Production of Interleukin 17 Induces Stem Cell Features of Pancreatic Intraepithelial Neoplasia Cells

Yu Zhang; Michelle Zoltan; Erick Riquelme; Hanwen Xu; Ismet Sahin; Susana Castro-Pando; Maria Fernanda Montiel; Kyle Chang; Zhengyu Jiang; Jianhua Ling; Sonal Gupta; William Horne; Melissa Pruski; Huamin Wang; Shao Cong Sun; Guillermina Lozano; Paul J. Chiao; Anirban Maitra; Steven D. Leach; Jay K. Kolls; Eduardo Vilar; Timothy C. Wang; Jennifer M. Bailey; Florencia McAllister

BACKGROUND & AIMS Little is known about how the immune system affects stem cell features of pancreatic cancer cells. Immune cells that produce interleukin 17A (IL17A) in the chronically inflamed pancreas (chronic pancreatitis) contribute to pancreatic interepithelial neoplasia (PanIN) initiation and progression. We investigated the effects that IL17A signaling exerts on pancreatic cancer progenitor cells and the clinical relevance of this phenomena. METHODS We performed studies with Mist1Cre;LSLKras;Rosa26mTmG (KCiMist;G) and Kras(G12D);Trp53(R172H);Pdx1-Cre (KPC) mice (which upon tamoxifen induction spontaneously develop PanINs) and control littermates. Some mice were injected with neutralizing antibodies against IL17A or control antibody. Pancreata were collected, PanIN epithelial cells were isolated by flow cytometry based on lineage tracing, and gene expression profiles were compared. We collected cells from pancreatic tumors of KPC mice, incubated them with IL17 or control media, measured expression of genes regulated by IL17 signaling, injected the cancer cells into immune competent mice, and measured tumor growth. IL17A was overexpressed in pancreata of KCiMist mice from an adenoviral vector. Pancreata were collected from all mice and analyzed by histology and immunohistochemistry. Levels of DCLK1 and other proteins were knocked down in KPC pancreatic cancer cells using small interfering or short hairpin RNAs; cells were analyzed by immunoblotting. We obtained 65 pancreatic tumor specimens from patients, analyzed protein levels by immunohistochemistry, and compared results with patient survival times. We also analyzed gene expression levels and patient outcome using The Cancer Genome Atlas database. RESULTS PanIN cells from KCiMist;G mice had a gene expression pattern associated with embryonic stem cells. Mice given injections of IL17-neutralizing antibodies, or with immune cells that did not secrete IL17, lost this expression pattern and had significantly decreased expression of DCLK1 and POU2F3, which regulate tuft cell development. KCiMist mice that overexpressed IL17 formed more PanINs, with more DCLK1-positive cells, than control mice. Pancreatic tumor cells from KPC mice and human Capan-2 cells exposed to IL17A had increased activation of NF-κB and mitogen-activated protein kinase signaling and increased expression of DCLK1 and ALDH1A1 (a marker of embryonic stem cells) compared with cells in control media. These cells also formed tumors faster that cells not exposed to IL17 when they were injected into immunocompetent mice. KPC cells with knockdown of DCLK1 expressed lower levels of ALDH1A1 after incubation with IL17 than cells without knockdown. Expression of the IL17 receptor C was higher in DCLK1-positive PanIN cells from mice compared with DCLK1-negative PanIN cells. In human pancreatic tumor tissues, high levels of DCLK1 associated with a shorter median survival time of patients (17.7 months, compared with 26.6 months of patients whose tumors had low levels of DCLK1). Tumor levels of POU2F3 and LAMC2 were also associated with patient survival time. CONCLUSIONS In studies of mouse and human pancreatic tumors and precursors, we found that immune cell-derived IL17 regulated development of tuft cells and stem cell features of pancreatic cancer cells via increased expression of DCLK1, POU2F3, ALDH1A1, and IL17RC. Strategies to disrupt this pathway might be developed to prevent pancreatic tumor growth and progression.


Cancer Research | 2017

Abstract LB-144: Tff2 labels pancreatic progenitors that lack proliferative potential during tissue regeneration but can serve as the origin of pancreatic cancer

Zhengyu Jiang; Bernhard W. Renz; Marina Macchini; Tanaka Takayuki; Ryota Takahashi; Giovanni Valenti; Woosook Kim; Wenju Chang; Yoku Hayakawa; Kosuke Sakitani; Moritz Middelhoff; Zinaida A. Dubeykovskaya; Timothy H. Chu; Karan Nagar; Yagnesh Tailor; Chythra R. Chandregowda; Akanksha Anand; Samuel Asfaha; Alina Iuga; Timothy C. Wang

While controversy over the existence of adult pancreatic stem cells persists, it is now appreciated that the acinar compartment of the pancreas harbors heterogeneous progenitors. Recent single-cell analysis also demonstrated the presence of molecularly distinct, albeit morphologically identical, acinar cell sub-lineages. Previously, using lineage-tracing approach, we reported the Dclk1+ facultative progenitors that are critical for pancreatic regeneration. Here, we identified a different pancreatic progenitor-like subpopulation which is labelled by trefoil factor 2 (Tff2), a known progenitor marker and capable of tracing multiple cell lineages in the stomach. In addition, Tff2 molecules have been shown to play a suppressive role in PDAC progression. We utilized constitutive Tff2Cre and inducible Tff2CreERT2-DTR mice which were generated through modification of a BAC allele. We crossed Tff2CreERT2-DTR with reporter mice (R26R-mTmG, -tdTomato) to trace Tff2 labeled cells, and found that Tff2 labels ~2 % of the overall population in the adult acinar compartment, which showed slow proliferation (1 year, descendants Citation Format: Zhengyu Jiang, Bernhard W. Renz, Marina Macchini, Tanaka Takayuki, Ryota Takahashi, Giovanni Valenti, Woosook Kim, Wenju Chang, Yoku Hayakawa, Kosuke Sakitani, Moritz Middelhoff, Zinaida Dubeykovskaya, Timothy Chu, Karan Nagar, Yagnesh Tailor, Chythra R. Chandregowda, Akanksha Anand, Samuel Asfaha, Alina C. Iuga, Timothy C. Wang. Tff2 labels pancreatic progenitors that lack proliferative potential during tissue regeneration but can serve as the origin of pancreatic cancer [abstract]. In: Proceedings of the American Association for Cancer Research Annual Meeting 2017; 2017 Apr 1-5; Washington, DC. Philadelphia (PA): AACR; Cancer Res 2017;77(13 Suppl):Abstract nr LB-144. doi:10.1158/1538-7445.AM2017-LB-144


Cancer Research | 2016

Abstract LB-272: Histidine decarboxylase (Hdc)-expressing myeloid cells support Foxp3+ Treg cells and promote colorectal cancer progression

Xiaowei Chen; Yoshihiro Takemoto; Karan Nagar; Timothy H. Chu; Zhengyu Jiang; Wenju Chang; Richard A. Friedman; Yagnesh Tailor; Daniel L. Worthley; Timothy C. Wang

The tumor microenvironment contains a diverse population of myeloid cells that are recruited from bone marrow and converted to immunosuppressive cells, thus mediating tumor cell escape from immune checkpoint. We have identified a subset of dynamic bone marrow myeloid cells, which can be identified by histidine decarboxylase (Hdc) mRNA and GFP expression in Hdc-GFP transgenic mice. Gene expression profiling showed that Hdc-GFP+ CD11b+Gr1+ myeloid cells express higher levels of cell cycle promoting genes such as Ccnd2, Ccnd3 and Cdc14a, while cell proliferation repressors including Cdc14b, Cdc16 and Cdk4 were downregulated in Hdc-GFP+ myeloid cells. To further elucidate the role of Hdc+ myeloid cells in tumor progression, we performed lineage-tracing studies using Hdc-creERT2;R26-LSL-TdTomato reporter mice. Pulsed with tamoxifen, the majority of TdTomato+ cells were localized initially to a group of CD11b+Gr1+ myeloid cells representing the highest Hdc mRNA expression in bone marrow, spleen and blood. Later on, TdTomato+CD11b+Gr1- F4/80+ macrophages can be detected, indicating a hierarchy of Hdc+ myeloid cells in which Hdc+CD11b+Gr1+ myeloid cells reside at the apex. In general, CD11b+ myeloid cells have a relative short lifespan ( In a mouse model of azoxymethane (AOM)/DSS colorectal carcinogenesis, Hdc-creERT2;R26-LSL-TdTomato;R26-LSL-DTA mice were injected with 10 mg/kg AOM and followed by 3 circles of 10 days 1.5% DSS ad libitum in drinking water. We found that Hdc-TdTomato labeled a proportion of tumor infiltrating CD11b+Gr1+ myeloid cells that expressed higher levels of Arg-1, Cox2, and Pdl1 transcripts. Continuous depletion of Hdc+ myeloid cells by administration of tamoxifen chow to induce DTA (diphtheria toxin subunit A) killing in Hdc-expressing myeloid cells abrogated half of the tumor-infiltrating MDSCs and released tumoricidal CD8+ T cells (> 15-fold), leading to decreased tumor number. This tumor suppression could be rescued by Hdc+ CD11b+Gr1+ cell adoptive transfer. Serum chemokine profiling revealed that Hdc+ DTA mediated myeloid depletion also decreased serum chemokine levels, among which Cxcl13 decreased the most (>30-fold). Cxcl13 protein and Cxcl13 mRNA also decreased in the colonic tumor tissue in the Hdc+ myeloid cell depleted AOM/DSS treatment group. Along with reduction in Cxcl13 levels, we also detected a significant reduction of Foxp3+ Treg cells in the tumor frozen sections stained with antibody against Foxp3 compared to R26-LSL-TdTomato;R26-LSL-DTA controls received the same treatments. Pre-knockdown of Cxcl13 by Dicer-substrate SiRNA (DsiRNAs) in a co-culture of splenic Hdc+ CD11b+Gr1+ myeloid cells from colon tumor-bearing mice with splenic Foxp3-GFP+ Treg cells induced apoptosis and decreased numbers of GFP+ cells compared to the scramble knockdown control group. Taken together, our data suggest that Hdc marks a distinct subset of myeloid cells with greater potency for promoting tumorigenicity, in part through supporting Tregs and suppressing CD8+ Tcells in the tumor microenvironment. Citation Format: Xiaowei Chen, Yoshihiro Takemoto, Karan K. Nagar, Timothy H. Chu, Zhengyu Jiang, Wenju Chang, Richard A. Friedman, Yagnesh H. Tailor, Daniel L. Worthley, Timothy C. Wang. Histidine decarboxylase (Hdc)-expressing myeloid cells support Foxp3+ Treg cells and promote colorectal cancer progression. [abstract]. In: Proceedings of the 107th Annual Meeting of the American Association for Cancer Research; 2016 Apr 16-20; New Orleans, LA. Philadelphia (PA): AACR; Cancer Res 2016;76(14 Suppl):Abstract nr LB-272.


Cancer Discovery | 2018

Cholinergic Signaling via Muscarinic Receptors Directly and Indirectly Suppresses Pancreatic Tumorigenesis and Cancer Stemness

Bernhard W. Renz; Takayuki Tanaka; Masaki Sunagawa; Ryota Takahashi; Zhengyu Jiang; Marina Macchini; Zahra Dantes; Giovanni Valenti; Ruth A. White; Moritz Middelhoff; Matthias Ilmer; Paul E. Oberstein; Martin K. Angele; Huan Deng; Yoku Hayakawa; C. Benedikt Westphalen; Jens Werner; Helen Remotti; Maximilian Reichert; Yagnesh Tailor; Karan Nagar; Richard A. Friedman; Alina C. Iuga; Kenneth P. Olive; Timothy C. Wang


Gastroenterology | 2017

Stromal Dclk1 Expression Labels Multi-Potential Neural Progenitor Cells in the Enteric Nervous System

Moritz Middelhoff; Yoku Hayakawa; Giovanni Valenti; Kosuke Sakitani; Ryota Takahashi; Takayuki Tanaka; Woosook Kim; Sama Sayin; Zinaida A. Dubeykovskaya; Zhengyu Jiang; Xiaowei Chen; Haibo Liu; Huan Deng; Yagnesh Tailor; Karan Nagar; Timothy H. Chu; Svetlana L. Sabel; Meenakshi Rao; Christoph B. Westphalen; Michael Quante; Kara Gross Margolis; Michael D. Gershon; Timothy C. Wang


Gastroenterology | 2016

803 MIST1 Positive Stem Cells in the Antrum Serve As a Cell-of-Origin for Gastric Cancer With APC Loss

Kosuke Sakitani; Yoku Hayakawa; Woosook Kim; Aleksandra M. Urbanska; Wenju Chang; Moritz Middelhoff; Zinaida A. Dubeykovskaya; Zhengyu Jiang; Takayuki Tanaka; Xiaowei Chen; Yagnesh Tailor; Karan Nagar; Timothy H. Chu; James G. Fox; Timothy C. Wang

Collaboration


Dive into the Zhengyu Jiang's collaboration.

Top Co-Authors

Avatar

Timothy C. Wang

Columbia University Medical Center

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Karan Nagar

Columbia University Medical Center

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Timothy H. Chu

Columbia University Medical Center

View shared research outputs
Top Co-Authors

Avatar

Moritz Middelhoff

Columbia University Medical Center

View shared research outputs
Top Co-Authors

Avatar

Richard A. Friedman

Columbia University Medical Center

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Giovanni Valenti

Columbia University Medical Center

View shared research outputs
Researchain Logo
Decentralizing Knowledge