Try a new search

Format these results:

Searched for:

person:wangj59

in-biosketch:yes

Total Results:

51


Myeloid dysregulation and therapeutic intervention in COVID-19

Gu, Runxia; Mao, Tianyang; Lu, Qiao; Tianjiao Su, Tina; Wang, Jun
The dysregulation of myeloid cell responses is increasingly demonstrated to be a major mechanism of pathogenesis for COVID-19. The pathological cellular and cytokine signatures associated with this disease point to a critical role of a hyperactivated innate immune response in driving pathology. Unique immunopathological features of COVID-19 include myeloid-cell dominant inflammation and cytokine release syndrome (CRS) alongside lymphopenia and acute respiratory distress syndrome (ARDS), all of which correlate with severe disease. Studies suggest a range of causes mediating myeloid hyperactivation, such as aberrant innate sensing, asynchronized immune cellular responses, as well as direct viral protein/host interactions. These include the recent identification of new myeloid cell receptors that bind SARS-CoV-2, which drive myeloid cell hyperinflammatory responses independently of lung epithelial cell infection via the canonical receptor, angiotensin-converting enzyme 2 (ACE2). The spectrum and nature of myeloid cell dysregulation in COVID-19 also differs from, at least to some extent, what is observed in other infectious diseases involving myeloid cell activation. While much of the therapeutic effort has focused on preventative measures with vaccines or neutralizing antibodies that block viral infection, recent clinical trials have also targeted myeloid cells and the associated cytokines as a means to resolve CRS and severe disease, with promising but thus far modest effects. In this review, we critically examine potential mechanisms driving myeloid cell dysregulation, leading to immunopathology and severe disease, and discuss potential therapeutic strategies targeting myeloid cells as a new paradigm for COVID-19 treatment.
PMCID:8576142
PMID: 34823995
ISSN: 1096-3618
CID: 5063812

Author Correction: Mechanical activation of spike fosters SARS-CoV-2 infection

Hu, Wei; Zhang, Yong; Fei, Panyu; Zhang, Tongtong; Yao, Danmei; Gao, Yufei; Liu, Jia; Chen, Hui; Lu, Qiao; Mudianto, Tenny; Zhang, Xinrui; Xiao, Chuxuan; Ye, Yang; Sun, Qiming; Zhang, Jing; Xie, Qi; Wang, Pei-Hui; Wang, Jun; Li, Zhenhai; Lou, Jizhong; Chen, Wei
PMID: 34588627
ISSN: 1748-7838
CID: 5067512

Mechanical activation of spike fosters SARS-CoV-2 viral infection

Hu, Wei; Zhang, Yong; Fei, Panyu; Zhang, Tongtong; Yao, Danmei; Gao, Yufei; Liu, Jia; Chen, Hui; Lu, Qiao; Mudianto, Tenny; Zhang, Xinrui; Xiao, Chuxuan; Ye, Yang; Sun, Qiming; Zhang, Jing; Xie, Qi; Wang, Pei-Hui; Wang, Jun; Li, Zhenhai; Lou, Jizhong; Chen, Wei
The outbreak of SARS-CoV-2 (SARS2) has caused a global COVID-19 pandemic. The spike protein of SARS2 (SARS2-S) recognizes host receptors, including ACE2, to initiate viral entry in a complex biomechanical environment. Here, we reveal that tensile force, generated by bending of the host cell membrane, strengthens spike recognition of ACE2 and accelerates the detachment of spike's S1 subunit from the S2 subunit to rapidly prime the viral fusion machinery. Mechanistically, such mechano-activation is fulfilled by force-induced opening and rotation of spike's receptor-binding domain to prolong the bond lifetime of spike/ACE2 binding, up to 4 times longer than that of SARS-S binding with ACE2 under 10 pN force application, and subsequently by force-accelerated S1/S2 detachment which is up to ~103 times faster than that in the no-force condition. Interestingly, the SARS2-S D614G mutant, a more infectious variant, shows 3-time stronger force-dependent ACE2 binding and 35-time faster force-induced S1/S2 detachment. We also reveal that an anti-S1/S2 non-RBD-blocking antibody that was derived from convalescent COVID-19 patients with potent neutralizing capability can reduce S1/S2 detachment by 3 × 106 times under force. Our study sheds light on the mechano-chemistry of spike activation and on developing a non-RBD-blocking but S1/S2-locking therapeutic strategy to prevent SARS2 invasion.
PMID: 34465913
ISSN: 1748-7838
CID: 5011712

Deep learning-based prediction of the T cell receptor-antigen binding specificity

Lu, Tianshi; Zhang, Ze; Zhu, James; Wang, Yunguan; Jiang, Peixin; Xiao, Xue; Bernatchez, Chantale; Heymach, John V; Gibbons, Don L; Wang, Jun; Xu, Lin; Reuben, Alexandre; Wang, Tao
Neoantigens play a key role in the recognition of tumor cells by T cells. However, only a small proportion of neoantigens truly elicit T cell responses, and fewer clues exist as to which neoantigens are recognized by which T cell receptors (TCRs). We built a transfer learning-based model, named pMHC-TCR binding prediction network (pMTnet), to predict TCR-binding specificities of neoantigens, and T cell antigens in general, presented by class I major histocompatibility complexes (pMHCs). pMTnet was comprehensively validated by a series of analyses, and showed advance over previous work by a large margin. By applying pMTnet in human tumor genomics data, we discovered that neoantigens were generally more immunogenic than self-antigens, but HERV-E, a special type of self-antigen that is re-activated in kidney cancer, is more immunogenic than neoantigens. We further discovered that patients with more clonally expanded T cells exhibiting better affinity against truncal, rather than subclonal, neoantigens, had more favorable prognosis and treatment response to immunotherapy, in melanoma and lung cancer but not in kidney cancer. Predicting TCR-neoantigen/antigen pairs is one of the most daunting challenges in modern immunology. However, we achieved an accurate prediction of the pairing only using the TCR sequence (CDR3β), antigen sequence, and class I MHC allele, and our work revealed unique insights into the interactions of TCRs and pMHCs in human tumors using pMTnet as a discovery tool.
PMCID:9396750
PMID: 36003885
ISSN: 2522-5839
CID: 5338312

A burned-out CD8+ T-cell subset expands in the tumor microenvironment and curbs cancer immunotherapy

Sanmamed, Miguel F; Nie, Xinxin; Desai, Shruti S; Villaroel-Espindola, Franz; Badri, Ti; Zhao, Dejian; Kim, Anthony W; Ji, Lan; Zhang, Tianxiang; Quinlan, Edward; Cheng, Xiaoxiao; Han, Xue; Vesely, Matthew D; Nassar, Ala F; Sun, Jingwei; Zhang, Yu; Kim, Tae Kon; Wang, Jun; Melero, Ignacio; Herbst, Roy S; Schalper, Kurt A; Chen, Lieping
Specific mechanisms by which tumor infiltrating lymphocytes (TIL) become dysfunctional remain poorly understood. Here, we employed a two-pronged approach using single-cell mass cytometry and tissue imaging technologies to dissect TILs from 25 resectable and 35 advanced non-small cell lung cancer (NSCLC) patients. We identified a burned-out CD8+ TIL subset (Ebo) that specifically accumulated within the tumor microenvironment (TME), but not in adjacent non-tumoral tissues. Ebo showed the highest expression of proliferation and activation markers, but produced the lowest amount of IFNy and were the most apoptotic CD8+ TIL subset. Using a humanized patient-derived tumor xenograft model, we demonstrated that Ebo expansion occurred within the TME in a PD-1/B7-H1 pathway-dependent manner. Ebo abundance in baseline tumor tissues was associated with resistance to anti-PD therapy in NSCLC patients. Our study identifies a dysfunctional TIL subset, with distinct features from previously described exhausted T cells, and implies strategies to overcome immunotherapy resistance.
PMID: 33658301
ISSN: 2159-8290
CID: 4819592

SARS-CoV-2 exacerbates proinflammatory responses in myeloid cells through C-type lectin receptors and Tweety family member 2

Lu, Qiao; Liu, Jia; Zhao, Shuai; Gomez Castro, Maria Florencia; Laurent-Rolle, Maudry; Dong, Jianbo; Ran, Xiaojuan; Damani-Yokota, Payal; Tang, Hongzhen; Karakousi, Triantafyllia; Son, Juhee; Kaczmarek, Maria E; Zhang, Ze; Yeung, Stephen T; McCune, Broc T; Chen, Rita E; Tang, Fei; Ren, Xianwen; Chen, Xufeng; Hsu, Jack C C; Teplova, Marianna; Huang, Betty; Deng, Haijing; Long, Zhilin; Mudianto, Tenny; Jin, Shumin; Lin, Peng; Du, Jasper; Zang, Ruochen; Su, Tina Tianjiao; Herrera, Alberto; Zhou, Ming; Yan, Renhong; Cui, Jia; Zhu, James; Zhou, Qiang; Wang, Tao; Ma, Jianzhu; Koralov, Sergei B; Zhang, Zemin; Aifantis, Iannis; Segal, Leopoldo N; Diamond, Michael S; Khanna, Kamal M; Stapleford, Kenneth A; Cresswell, Peter; Liu, Yue; Ding, Siyuan; Xie, Qi; Wang, Jun
Despite mounting evidence of severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) engagement with immune cells, most express little, if any, of the canonical receptor of SARS-CoV-2, angiotensin-converting enzyme 2 (ACE2). Here, using a myeloid cell receptor-focused ectopic expression screen, we identified several C-type lectins (DC-SIGN, L-SIGN, LSECtin, ASGR1, and CLEC10A) and Tweety family member 2 (TTYH2) as glycan-dependent binding partners of the SARS-CoV-2 spike. Except for TTYH2, these molecules primarily interacted with spike via regions outside of the receptor-binding domain. Single-cell RNA sequencing analysis of pulmonary cells from individuals with coronavirus disease 2019 (COVID-19) indicated predominant expression of these molecules on myeloid cells. Although these receptors do not support active replication of SARS-CoV-2, their engagement with the virus induced robust proinflammatory responses in myeloid cells that correlated with COVID-19 severity. We also generated a bispecific anti-spike nanobody that not only blocked ACE2-mediated infection but also the myeloid receptor-mediated proinflammatory responses. Our findings suggest that SARS-CoV-2-myeloid receptor interactions promote immune hyperactivation, which represents potential targets for COVID-19 therapy.
PMID: 34048708
ISSN: 1097-4180
CID: 4888442

Prevalence and associated risk factors of Helicobacter pylori infection in the Wuwei cohort of north-western China

Zhang, Fuhua; Pu, Ke; Wu, Zhengqi; Zhang, Zhiyi; Liu, Xin; Chen, Zhaofeng; Ye, Yuwei; Wang, Yuping; Zheng, Ya; Zhang, Jinhua; An, Feng; Zhao, Shijun; Hu, Xiaobin; Li, Youpeng; Li, Qiang; Liu, Min; Lu, Hong; Zhang, Hongling; Zhao, Yue; Yuan, Hao; Ding, Xiangping; Shu, Xiaochuang; Ren, Qian; Gou, Xi; Hu, Zenan; Wang, Jun; Wang, Yuling; Guan, Quanlin; Guo, Qinghong; Ji, Rui; Zhou, Yongning
OBJECTIVES:To evaluate the prevalence of Helicobacter pylori infection and risk factors and to serotype the strains in Wuwei, located in north-western China, which has a high incidence of gastric cancer. METHODS:C-urea breath test, and H. pylori antibody were detected in 9183 serum samples by latex immunoturbidimetric method. The correlation of H. pylori infection with demographic-economic, lifestyle factors and medical history among the participants was determined by questionnaire. The antibodies against H. pylori urease, VacA and CagA in serum were determined by dot immunobinding assay. RESULTS:The infection rate of H. pylori was 53.0%, and 90.1% of strains were type I strains. The H. pylori infection rate was higher among farmers (OR = 1.34, 95% CI: 1.19-1.50) and individuals who had a junior high school or higher education level (OR = 1.10, 95% CI: 1.06-1.15), and was lower in older individuals (OR = 0.86, 95% CI: 0.83-0.90), individuals with high income (OR = 0.93, 95% CI: 0.90-0.95), individuals with a habit of eating quickly (OR = 0.93, 95% CI: 0.87-0.99) and individuals who consumed more fruit and vegetables (OR = 0.90, 95% CI: 0.85-0.95). Individuals with history of cholecystitis/cholecystolithiasis, hypertension and asthma were negatively correlated with H. pylori infection (P < 0.05). CONCLUSION:The prevalence of H. pylori infection is high in Wuwei. The major prevalent strain is type I strain. Age, education, occupation, household income, consumption of fruit and vegetables, and habit of eating quickly are independent risk factors for H. pylori infection, which is also associated with individuals with a history of extragastric diseases.
PMID: 33159827
ISSN: 1365-3156
CID: 4920552

Siglec-15 as an Emerging Target for Next-generation Cancer Immunotherapy

Sun, Jingwei; Lu, Qiao; Sanmanmed, Miguel F; Wang, Jun
Immunomodulatory agents blocking the PD-1/PD-L1 pathway have shown a new way to treat cancer. The explanation underlying the success of these agents may be the selective expression of PD-L1 with dominant immune-suppressive activities in the tumor microenvironment (TME), supporting a more favorable tumor response-to-toxicity ratio. However, despite the big success of these drugs, most patients with cancer show primary or acquired resistance, calling for the identification of new immune modulators in the TME. Using a genome-scale T-cell activity array in combination with bioinformatic analysis of human cancer databases, we identified Siglec-15 as a critical immune suppressor with broad upregulation on various cancer types and a potential target for cancer immunotherapy. Siglec-15 has unique molecular features compared with many other known checkpoint inhibitory ligands. It shows prominent expression on macrophages and cancer cells and a mutually exclusive expression with PD-L1, suggesting that it may be a critical immune evasion mechanism in PD-L1-negative patients. Interestingly, Siglec-15 has also been identified as a key regulator for osteoclast differentiation and may have potential implications in bone disorders not limited to osteoporosis. Here, we provide an overview of Siglec-15 biology, its role in cancer immune regulation, the preliminary and encouraging clinical data related to the first-in-class Siglec-15 targeting mAb, as well as many unsolved questions in this pathway. As a new player in the cancer immunotherapeutic arena, Siglec-15 may represent a novel class of immune inhibitors with tumor-associated expression and divergent mechanisms of action to PD-L1, with potential implications in anti-PD-1/PD-L1-resistant patients.
PMID: 32958700
ISSN: 1557-3265
CID: 4789592

The RNA helicase Dhx15 mediates Wnt-induced antimicrobial protein expression in Paneth cells

Wang, Yalong; He, Kaixin; Sheng, Baifa; Lei, Xuqiu; Tao, Wanyin; Zhu, Xiaoliang; Wei, Zheng; Fu, Rongjie; Wang, Anlei; Bai, Shengdan; Zhang, Zhao; Hong, Na; Ye, Chao; Tian, Ye; Wang, Jun; Li, Mingsong; Zhang, Kaiguang; Li, Lin; Yang, Hua; Li, Hua-Bing; Flavell, Richard A; Zhu, Shu
RNA helicases play roles in various essential biological processes such as RNA splicing and editing. Recent in vitro studies show that RNA helicases are involved in immune responses toward viruses, serving as viral RNA sensors or immune signaling adaptors. However, there is still a lack of in vivo data to support the tissue- or cell-specific function of RNA helicases owing to the lethality of mice with complete knockout of RNA helicases; further, there is a lack of evidence about the antibacterial role of helicases. Here, we investigated the in vivo role of Dhx15 in intestinal antibacterial responses by generating mice that were intestinal epithelial cell (IEC)-specific deficient for Dhx15 (Dhx15 f/f Villin1-cre, Dhx15ΔIEC). These mice are susceptible to infection with enteric bacteria Citrobacter rodentium (C. rod), owing to impaired α-defensin production by Paneth cells. Moreover, mice with Paneth cell-specific depletion of Dhx15 (Dhx15 f/f Defensinα6-cre, Dhx15ΔPaneth) are more susceptible to DSS (dextran sodium sulfate)-induced colitis, which phenocopy Dhx15ΔIEC mice, due to the dysbiosis of the intestinal microbiota. In humans, reduced protein levels of Dhx15 are found in ulcerative colitis (UC) patients. Taken together, our findings identify a key regulator of Wnt-induced α-defensins in Paneth cells and offer insights into its role in the antimicrobial response as well as intestinal inflammation.
PMID: 33483420
ISSN: 1091-6490
CID: 4767862

A modulator of the low-voltage-activated T-type calcium channel that reverses HIV glycoprotein 120-, paclitaxel-, and spinal nerve ligation-induced peripheral neuropathies

Cai, Song; Tuohy, Peter; Ma, Chunlong; Kitamura, Naoya; Gomez, Kimberly; Zhou, Yuan; Ran, Dongzhi; Bellampalli, Shreya Sai; Yu, Jie; Luo, Shizhen; Dorame, Angie; Yen Ngan Pham, Nancy; Molnar, Gabriella; Streicher, John M; Patek, Marcel; Perez-Miller, Samantha; Moutal, Aubin; Wang, Jun; Khanna, Rajesh
The voltage-gated calcium channels CaV3.1-3.3 constitute the T-type subfamily, whose dysfunctions are associated with epilepsy, psychiatric disorders, and chronic pain. The unique properties of low-voltage-activation, faster inactivation, and slower deactivation of these channels support their role in modulation of cellular excitability and low-threshold firing. Thus, selective T-type calcium channel antagonists are highly sought after. Here, we explored Ugi-azide multicomponent reaction products to identify compounds targeting T-type calcium channel. Of the 46 compounds tested, an analog of benzimidazolonepiperidine-5bk (1-{1-[(R)-{1-[(1S)-1-phenylethyl]-1H-1,2,3,4-tetrazol-5-yl}(thiophen-3-yl)methyl]piperidin-4-yl}-2,3-dihydro-1H-1,3-benzodiazol-2-one) modulated depolarization-induced calcium influx in rat sensory neurons. Modulation of T-type calcium channels by 5bk was further confirmed in whole-cell patch clamp assays in dorsal root ganglion (DRG) neurons, where pharmacological isolation of T-type currents led to a time- and concentration-dependent regulation with a low micromolar IC50. Lack of an acute effect of 5bk argues against a direct action on T-type channels. Genetic knockdown revealed CaV3.2 to be the isoform preferentially modulated by 5bk. High voltage-gated calcium, as well as tetrodotoxin-sensitive and -resistant sodium, channels were unaffected by 5bk. 5bk inhibited spontaneous excitatory postsynaptic currents and depolarization-evoked release of calcitonin gene-related peptide from lumbar spinal cord slices. Notably, 5bk did not bind human mu, delta, or kappa opioid receptors. 5bk reversed mechanical allodynia in rat models of HIV-associated neuropathy, chemotherapy-induced peripheral neuropathy, and spinal nerve ligation-induced neuropathy, without effects on locomotion or anxiety. Thus, 5bk represents a novel T-type modulator that could be used to develop nonaddictive pain therapeutics.
PMCID:7572723
PMID: 32541387
ISSN: 1872-6623
CID: 5121352