Network


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

Hotspot


Dive into the research topics where Stephen Chiu is active.

Publication


Featured researches published by Stephen Chiu.


Journal of Experimental Medicine | 2017

Monocyte-derived alveolar macrophages drive lung fibrosis and persist in the lung over the life span.

Alexander V. Misharin; Luisa Morales-Nebreda; Paul A. Reyfman; Carla M. Cuda; James M. Walter; Alexandra C. McQuattie-Pimentel; C-I Chen; Kishore R. Anekalla; N Joshi; Kjn Williams; Hiam Abdala-Valencia; Tj Yacoub; Monica Chi; Stephen Chiu; Francisco J. Gonzalez-Gonzalez; Khalilah L. Gates; Anna P. Lam; Trevor T. Nicholson; Philip J. Homan; Saul Soberanes; Salina Dominguez; Vk Morgan; Rana Saber; Alexander M. Shaffer; Monique Hinchcliff; Stacy A. Marshall; Ankit Bharat; Sergejs Berdnikovs; Sangeeta Bhorade; Elizabeth Bartom

Little is known about the relative importance of monocyte and tissue-resident macrophages in the development of lung fibrosis. We show that specific genetic deletion of monocyte-derived alveolar macrophages after their recruitment to the lung ameliorated lung fibrosis, whereas tissue-resident alveolar macrophages did not contribute to fibrosis. Using transcriptomic profiling of flow-sorted cells, we found that monocyte to alveolar macrophage differentiation unfolds continuously over the course of fibrosis and its resolution. During the fibrotic phase, monocyte-derived alveolar macrophages differ significantly from tissue-resident alveolar macrophages in their expression of profibrotic genes. A population of monocyte-derived alveolar macrophages persisted in the lung for one year after the resolution of fibrosis, where they became increasingly similar to tissue-resident alveolar macrophages. Human homologues of profibrotic genes expressed by mouse monocyte-derived alveolar macrophages during fibrosis were up-regulated in human alveolar macrophages from fibrotic compared with normal lungs. Our findings suggest that selectively targeting alveolar macrophage differentiation within the lung may ameliorate fibrosis without the adverse consequences associated with global monocyte or tissue-resident alveolar macrophage depletion.


Science Translational Medicine | 2017

Donor pulmonary intravascular nonclassical monocytes recruit recipient neutrophils and mediate primary lung allograft dysfunction

Zhikun Zheng; Stephen Chiu; Mahzad Akbarpour; Haiying Sun; Paul A. Reyfman; Kishore R. Anekalla; Hiam Abdala-Valencia; Daphne Edgren; Wenjun Li; Daniel Kreisel; Farida Korobova; Ramiro Fernandez; Alexandra C. McQuattie-Pimentel; Zheng Zhang; Harris Perlman; Alexander V. Misharin; G. R. Scott Budinger; Ankit Bharat

Donor nonclassical monocytes mediate primary lung allograft dysfunction by recruiting neutrophils via MyD88-dependent production of CXCL2. Nonclassical monocytes prompt primary graft dysfunction Despite concerted efforts, primary graft dysfunction is a major cause of graft failure after organ transplantation. In lung transplantation, primary graft dysfunction is known to be mediated by early neutrophil infiltration. Zheng et al. used syngeneic and allogeneic mouse models of lung transplantation to show that nonclassical monocytes were the key cell population recruiting these destructive neutrophils. These intravascular cells were donor-derived and were also detectable in human lung grafts being used for transplant. Because depletion of nonclassical monocytes prevented primary graft dysfunction in the mouse models, targeting this cell population during human transplant could lead to improved rates of graft success. Primary graft dysfunction is the predominant driver of mortality and graft loss after lung transplantation. Recruitment of neutrophils as a result of ischemia-reperfusion injury is thought to cause primary graft dysfunction; however, the mechanisms that regulate neutrophil influx into the injured lung are incompletely understood. We found that donor-derived intravascular nonclassical monocytes (NCMs) are retained in human and murine donor lungs used in transplantation and can be visualized at sites of endothelial injury after reperfusion. When NCMs in the donor lungs were depleted, either pharmacologically or genetically, neutrophil influx and lung graft injury were attenuated in both allogeneic and syngeneic models. Similar protection was observed when the patrolling function of donor NCMs was impaired by deletion of the fractalkine receptor CX3CR1. Unbiased transcriptomic profiling revealed up-regulation of MyD88 pathway genes and a key neutrophil chemoattractant, CXCL2, in donor-derived NCMs after reperfusion. Reconstitution of NCM-depleted donor lungs with wild-type but not MyD88-deficient NCMs rescued neutrophil migration. Donor NCMs, through MyD88 signaling, were responsible for CXCL2 production in the allograft and neutralization of CXCL2 attenuated neutrophil influx. These findings suggest that therapies to deplete or inhibit NCMs in donor lung might ameliorate primary graft dysfunction with minimal toxicity to the recipient.


Current Opinion in Organ Transplantation | 2016

Role of monocytes and macrophages in regulating immune response following lung transplantation.

Stephen Chiu; Ankit Bharat

Purpose of reviewAdvances in the field of monocyte and macrophage biology have dramatically changed our understanding of their role during homeostasis and inflammation. Here we review the role of these important innate immune effectors in the lung during inflammatory challenges including lung transplantation. Recent findingsNeutrophil extravasation into lung tissue and the alveolar space have been shown to be pathogenic during acute lung injury as well as primary graft dysfunction following lung transplantation. Recent advances in lung immunology have demonstrated the remarkable plasticity of both monocytes and macrophages and demonstrated their importance as mediators of neutrophil recruitment and transendothelial migration during inflammation. SummaryMonocytes and macrophages are emerging as key players in mediating both the pathogen response and sterile lung inflammation, including that arising from barotrauma and ischemia-reperfusion injury. Ongoing studies will establish the mechanisms by which these monocytes and macrophages initiate a variety of immune response that lay the fundamental basis of injury response in the lung.


Surgery | 2015

Decreased CXCL12 is associated with impaired alveolar epithelial cell migration and poor lung healing after lung resection.

Jacob Kanter; Haiying Sun; Stephen Chiu; Malcolm M. DeCamp; Peter H. S. Sporn; Jacob I. Sznajder; Ankit Bharat

BACKGROUND Prolonged air leak (PAL) is an important cause of morbidity and mortality after lung resection, but its pathogenesis has not been elucidated. Migration of alveolar type II epithelial cells is essential for lung wound repair. Here we determined the role of C-X-C motif chemokine 12 (CXCL12) on alveolar epithelial cell migration and lung wound healing. METHODS CXCL12 in the pleural fluid of patients was analyzed using enzyme-linked immunosorbent assay. Human A549 and murine MLE12 alveolar epithelial cell lines were used for wound closure, cell migration, and proliferation assays. Western blot was used to analyze Rac1 and cofilin. RESULTS Pleural CXCL12 was decreased in patients with PAL (1,389 ± 192 vs 3,270 ± 247 pg/mL; P < .0001). CXCL12 enhanced scratch wound closure in both A549 (77.9 ± 0.7% vs 71.5 ± 0.4%; P = .0016) and MLE12 (92.9 ± 4.9% vs 66.0 ± 4.8%; P = .017). CXCL12 enhanced migration by 57% in A549 (P = .0008) and by 86% in MLE12 (P < .0001). AMD3100, a selective CXCR4 antagonist, prevented the effects of CXCL12. CXCL12 increased Rac1 and cofilin activation but did not change bromodeoxyuridine incorporation or cell counts. CONCLUSION Reduced pleural CXCL12 is associated with PAL. CXCL12 promotes alveolar epithelial cell migration by binding to its receptor CXCR4 and may have a role in lung healing. CXCL12-mediated alveolar epithelial cell migration is associated with Rac1 and cofilin activation.


The Annals of Thoracic Surgery | 2016

Humoral Human Lung Allograft Rejection by Tissue-Restricted Non-HLA Antibodies

Ramiro Fernandez; Stephen Chiu; Kirtee Raparia; Puneet Garcha; Carol Farver; Marie Budev; Anat R. Tambur; Malcolm M. DeCamp; Scott Budinger; Harris Perlman; T. Mohanakumar; Ankit Bharat

A third of lung recipients have preexisting antibodies against nonhuman leukocyte self-antigens (nHAbs) present in the lung tissue. These nHAbs also form de novo in about 70% of patients within 3 years after transplantation. Both preexisting and de novo nHAbs can cause murine lung allograft dysfunction. However, their role in human transplantation remains unclear. We report hyperacute rejection after right lung transplant in a recipient with preexisting nHAbs. The recipient of the left lung from the same donor had an uneventful initial course, but de novo nHAbs developed at 3 weeks, leading to acute humoral rejection. Both patients were successfully treated with antibody-directed therapies.


Journal of Immunology | 2016

Lung Injury Combined with Loss of Regulatory T Cells Leads to De Novo Lung-Restricted Autoimmunity

Stephen Chiu; Ramiro Fernandez; Vijay Subramanian; Haiying Sun; Malcolm M. DeCamp; Daniel Kreisel; Harris Perlman; G. R. Scott Budinger; Thalachallour Mohanakumar; Ankit Bharat

More than one third of patients with chronic lung disease undergoing lung transplantation have pre-existing Abs against lung-restricted self-Ags, collagen type V (ColV), and k-α1 tubulin (KAT). These Abs can also develop de novo after lung transplantation and mediate allograft rejection. However, the mechanisms leading to lung-restricted autoimmunity remain unknown. Because these self-Ags are normally sequestered, tissue injury is required to expose them to the immune system. We previously showed that respiratory viruses can induce apoptosis in CD4+CD25+Foxp3+ regulatory T cells (Tregs), the key mediators of self-tolerance. Therefore, we hypothesized that lung-tissue injury can lead to lung-restricted immunity if it occurs in a setting when Tregs are impaired. We found that human lung recipients who suffer respiratory viral infections experienced a decrease in peripheral Tregs. Pre-existing lung allograft injury from donor-directed Abs or gastroesophageal reflux led to new ColV and KAT Abs post respiratory viral infection. Similarly, murine parainfluenza (Sendai) respiratory viral infection caused a decrease in Tregs. Intratracheal instillation of anti-MHC class I Abs, but not isotype control, followed by murine Sendai virus infection led to development of Abs against ColV and KAT, but not collagen type II (ColII), a cartilaginous protein. This was associated with expansion of IFN-γ–producing CD4+ T cells specific to ColV and KAT, but not ColII. Intratracheal anti-MHC class I Abs or hydrochloric acid in Foxp3-DTR mice induced ColV and KAT, but not ColII, immunity, only if Tregs were depleted using diphtheria toxin. We conclude that tissue injury combined with loss of Tregs can lead to lung-tissue–restricted immunity.


Current Transplantation Reports | 2015

Effects of Hypercapnia in Lung Tissue Repair and Transplant

Stephen Chiu; Jacob Kanter; Haiying Sun; Anchit Bharat; Peter H. S. Sporn; Ankit Bharat

Mammalian cells sense and transduce signals in response to high levels of carbon dioxide. Hypercapnia has a variety of effects on epithelial and immune cells which can be beneficial or detrimental depending on the biologic context. For instance, hypercapnia-mediated suppression of cell proliferation and migration can delay wound repair. Similarly, suppression of macrophages and the inflammatory response during hypercapnia can limit the host’s ability to clear pathogens. However, the suppressive effects of hypercapnia on immunity have potential benefits in the setting of transplantation. Here, we discuss the effects of high levels of carbon dioxide on lung healing and the potential applications in lung transplantation.


bioRxiv | 2018

Single-Cell Transcriptomic Analysis of Human Lung Reveals Complex Multicellular Changes During Pulmonary Fibrosis

Paul A. Reyfman; James M. Walter; Nikita Joshi; Kishore R. Anekalla; Alexandra C. McQuattie-Pimentel; Stephen Chiu; Ramiro Fernandez; Mahzad Akbarpour; Ching-I Chen; Ziyou Ren; Rohan Verma; Hiam Abdala-Valencia; Kiwon Nam; Monica Chi; SeungHye Han; Francisco J. Gonzalez-Gonzalez; Saul Soberanes; Satoshi Watanabe; Kinola J.N. Williams; Annette S. Flozak; Trevor T. Nicholson; Vk Morgan; Cara L. Hrusch; Robert D. Guzy; Catherine A. Bonham; Anne I. Sperling; Remzi Bag; Robert B. Hamanaka; Gökhan M. Mutlu; Anjana V. Yeldandi

Pulmonary fibrosis is a devastating disorder that results in the progressive replacement of normal lung tissue with fibrotic scar. Available therapies slow disease progression, but most patients go on to die or require lung transplantation. Single-cell RNA-seq is a powerful tool that can reveal cellular identity via analysis of the transcriptome, but its ability to provide biologically or clinically meaningful insights in a disease context is largely unexplored. Accordingly, we performed single-cell RNA-seq on lung tissue obtained from eight transplant donors and eight recipients with pulmonary fibrosis and one bronchoscopic cryobiospy sample. Integrated single-cell transcriptomic analysis of donors and patients with pulmonary fibrosis identified the emergence of distinct populations of epithelial cells and macrophages that were common to all patients with lung fibrosis. Analysis of transcripts in the Wnt pathway suggested that within the same cell type, Wnt secretion and response are restricted to distinct non-overlapping cells, which was confirmed using in situ RNA hybridization. Single-cell RNA-seq revealed heterogeneity within alveolar macrophages from individual patients, which was confirmed by immunohistochemistry. These results support the feasibility of discovery-based approaches applying next generation sequencing technologies to clinically obtained samples with a goal of developing personalized therapies. One Sentence Summary Single-cell RNA-seq applied to tissue from diseased and donor lungs and a living patient with pulmonary fibrosis identifies cell type-specific disease-associated molecular pathways.


Journal of Clinical Investigation | 2018

Spleen-derived classical monocytes mediate lung ischemia-reperfusion injury through IL-1β

Hsi-Min Hsiao; Ramiro Fernandez; Satona Tanaka; Wenjun Li; Jessica H. Spahn; Stephen Chiu; Mahzad Akbarpour; Daniel Ruiz-Pérez; Qiang Wu; Cem Turam; Davide Scozzi; Tsuyoshi Takahashi; Hannah Luehmann; Varun Puri; G. R. Scott Budinger; Alexander S. Krupnick; Alexander V. Misharin; Kory J. Lavine; Yongjian Liu; Andrew Gelman; Ankit Bharat; Daniel Kreisel

&NA; Ischemia‐reperfusion injury, a form of sterile inflammation, is the leading risk factor for both short‐term mortality following pulmonary transplantation and chronic lung allograft dysfunction. While it is well recognized that neutrophils are critical mediators of acute lung injury, processes that guide their entry into pulmonary tissue are not well understood. Here, we found that CCR2+ classical monocytes are necessary and sufficient for mediating extravasation of neutrophils into pulmonary tissue during ischemia‐reperfusion injury following hilar clamping or lung transplantation. The classical monocytes were mobilized from the host spleen, and splenectomy attenuated the recruitment of classical monocytes as well as the entry of neutrophils into injured lung tissue, which was associated with improved graft function. Neutrophil extravasation was mediated by MyD88‐dependent IL‐1&bgr; production by graft‐infiltrating classical monocytes, which downregulated the expression of the tight junction‐associated protein ZO‐2 in pulmonary vascular endothelial cells. Thus, we have uncovered a crucial role for classical monocytes, mobilized from the spleen, in mediating neutrophil extravasation, with potential implications for targeting of recipient classical monocytes to ameliorate pulmonary ischemia‐reperfusion injury in the clinic.


Cell Metabolism | 2018

Metformin Targets Mitochondrial Electron Transport to Reduce Air-Pollution-Induced Thrombosis.

Saul Soberanes; Alexander V. Misharin; Amit Jairaman; Luisa Morales-Nebreda; Alexandra C. McQuattie-Pimentel; Takugo Cho; Robert B. Hamanaka; Angelo Y. Meliton; James M. Walter; Ching-I Chen; Monica Chi; Stephen Chiu; Francisco J. Gonzalez-Gonzalez; Matthew Antalek; Hiam Adbala-Valencia; Sergio E. Chiarella; Kaitlyn Sun; Parker S. Woods; Andrew J. Ghio; Manu Jain; Harris Perlman; Karen M. Ridge; Richard I. Morimoto; Jacob I. Sznajder; William E. Balch; Sangeeta Bhorade; Ankit Bharat; Murali Prakriya; Navdeep S. Chandel; Gökhan M. Mutlu

Urban particulate matter air pollution induces the release of pro-inflammatory cytokines including interleukin-6 (IL-6) from alveolar macrophages, resulting in an increase in thrombosis. Here, we report that metformin provides protection in this murine model. Treatment of mice with metformin or exposure of murine or human alveolar macrophages to metformin prevented the particulate matter-induced generation of complex III mitochondrial reactive oxygen species, which were necessary for the opening of calcium release-activated channels (CRAC) and release of IL-6. Targeted genetic deletion of electron transport or CRAC channels in alveolar macrophages in mice prevented particulate matter-induced acceleration of arterial thrombosis. These findings suggest metformin as a potential therapy to prevent some of the premature deaths attributable to air pollution exposure worldwide.

Collaboration


Dive into the Stephen Chiu's collaboration.

Top Co-Authors

Avatar

Ankit Bharat

Northwestern University

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Daniel Kreisel

Washington University in St. Louis

View shared research outputs
Top Co-Authors

Avatar

Haiying Sun

Northwestern University

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Researchain Logo
Decentralizing Knowledge