274 results on '"Harold A. Chapman"'
Search Results
2. Alveolar regeneration through a Krt8+ transitional stem cell state that persists in human lung fibrosis
- Author
-
Maximilian Strunz, Lukas M. Simon, Meshal Ansari, Jaymin J. Kathiriya, Ilias Angelidis, Christoph H. Mayr, George Tsidiridis, Marius Lange, Laura F. Mattner, Min Yee, Paulina Ogar, Arunima Sengupta, Igor Kukhtevich, Robert Schneider, Zhongming Zhao, Carola Voss, Tobias Stoeger, Jens H. L. Neumann, Anne Hilgendorff, Jürgen Behr, Michael O’Reilly, Mareike Lehmann, Gerald Burgstaller, Melanie Königshoff, Harold A. Chapman, Fabian J. Theis, and Herbert B. Schiller
- Subjects
Science - Abstract
Injury repair is characterized by the generation of transient cell states important for tissue recovery. Here, the authors present a single cell RNA-seq map of recovery from bleomycin lung injury in mice and uncover a Krt8+ transitional stem cell state that precedes the regeneration of AT1 cells and persists in human lung fibrosis.
- Published
- 2020
- Full Text
- View/download PDF
3. Contextual cues from cancer cells govern cancer-associated fibroblast heterogeneity
- Author
-
Neus Bota-Rabassedas, Priyam Banerjee, Yichi Niu, Wenjian Cao, Jiayi Luo, Yuanxin Xi, Xiaochao Tan, Kuanwei Sheng, Young-Ho Ahn, Sieun Lee, Edwin Roger Parra, Jaime Rodriguez-Canales, Jacob Albritton, Michael Weiger, Xin Liu, Hou-Fu Guo, Jiang Yu, B. Leticia Rodriguez, Joshua J.A. Firestone, Barbara Mino, Chad J. Creighton, Luisa M. Solis, Pamela Villalobos, Maria Gabriela Raso, Daniel W. Sazer, Don L. Gibbons, William K. Russell, Gregory D. Longmore, Ignacio I. Wistuba, Jing Wang, Harold A. Chapman, Jordan S. Miller, Chenghang Zong, and Jonathan M. Kurie
- Subjects
cancer-associated fibroblast ,EMT ,metastasis ,lung cancer ,tumor microenvironment ,microRNA ,Biology (General) ,QH301-705.5 - Abstract
Summary: Cancer cells function as primary architects of the tumor microenvironment. However, the molecular features of cancer cells that govern stromal cell phenotypes remain unclear. Here, we show that cancer-associated fibroblast (CAF) heterogeneity is driven by lung adenocarcinoma (LUAD) cells at either end of the epithelial-to-mesenchymal transition (EMT) spectrum. LUAD cells that have high expression of the EMT-activating transcription factor ZEB1 reprogram CAFs through a ZEB1-dependent secretory program and direct CAFs to the tips of invasive projections through a ZEB1-driven CAF repulsion process. The EMT, in turn, sensitizes LUAD cells to pro-metastatic signals from CAFs. Thus, CAFs respond to contextual cues from LUAD cells to promote metastasis.
- Published
- 2021
- Full Text
- View/download PDF
4. Supplementary Table 1, Figures 1-9, Methods from Suppression of Tumor Invasion and Metastasis by Concurrent Inhibition of c-Met and VEGF Signaling in Pancreatic Neuroendocrine Tumors
- Author
-
Donald M. McDonald, Dana T. Aftab, James G. Christensen, Harold A. Chapman, Weon-Kyoo You, Sebastien P. Tabruyn, Vikash Bhagwandin, Casey W. Williamson, Ryan M. Naylor, Ying Wei, Toshina Ishiguro-Oonuma, and Barbara Sennino
- Abstract
PDF file - 2MB
- Published
- 2023
- Full Text
- View/download PDF
5. T Cells from NOD-PerIg Mice Target Both Pancreatic and Neuronal Tissue
- Author
-
Rachel Ettinger, David V. Serreze, Torrian Green, Zoë Bichler, Timothy J. Hines, Jeremy J. Racine, Brynn M Cairns, Laura C. Anderson, Abigail L. D. Tadenev, Rosalinda Doty, Jacqueline K. White, Robert W. Burgess, Janine M Wotton, Harold D. Chapman, and Meaghan E Dyer
- Subjects
Transgene ,Immunology ,Neuritis ,Peripherin ,Spleen ,Nod ,Biology ,Molecular biology ,medicine.anatomical_structure ,Splenocyte ,medicine ,Immunology and Allergy ,Peripheral neuritis ,CD8 - Abstract
It has become increasingly appreciated that autoimmune responses against neuronal components play an important role in type 1 diabetes (T1D) pathogenesis. In fact, a large proportion of islet-infiltrating B lymphocytes in the NOD mouse model of T1D produce Abs directed against the neuronal type III intermediate filament protein peripherin. NOD-PerIg mice are a previously developed BCR-transgenic model in which virtually all B lymphocytes express the H and L chain Ig molecules from the intra-islet–derived anti-peripherin–reactive hybridoma H280. NOD-PerIg mice have accelerated T1D development, and PerIg B lymphocytes actively proliferate within islets and expand cognitively interactive pathogenic T cells from a pool of naive precursors. We now report adoptively transferred T cells or whole splenocytes from NOD-PerIg mice expectedly induce T1D in NOD.scid recipients but, depending on the kinetics of disease development, can also elicit a peripheral neuritis (with secondary myositis). This neuritis was predominantly composed of CD4+ and CD8+ T cells. Ab depletion studies showed neuritis still developed in the absence of NOD-PerIg CD8+ T cells but required CD4+ T cells. Surprisingly, sciatic nerve–infiltrating CD4+ cells had an expansion of IFN-γ− and TNF-α− double-negative cells compared with those within both islets and spleen. Nerve and islet-infiltrating CD4+ T cells also differed by expression patterns of CD95, PD-1, and Tim-3. Further studies found transitory early B lymphocyte depletion delayed T1D onset in a portion of NOD-PerIg mice, allowing them to survive long enough to develop neuritis outside of the transfer setting. Together, this study presents a new model of peripherin-reactive B lymphocyte–dependent autoimmune neuritis.
- Published
- 2020
- Full Text
- View/download PDF
6. Human alveolar type 2 epithelium transdifferentiates into metaplastic KRT5+ basal cells
- Author
-
Jaymin J. Kathiriya, Chaoqun Wang, Minqi Zhou, Alexis Brumwell, Monica Cassandras, Claude Jourdan Le Saux, Max Cohen, Kostantinos-Dionysios Alysandratos, Bruce Wang, Paul Wolters, Michael Matthay, Darrell N. Kotton, Harold A. Chapman, and Tien Peng
- Subjects
Cells ,Respiratory Mucosa ,Inbred C57BL ,SCID ,Autoimmune Disease ,Medical and Health Sciences ,Transgenic ,Mesoderm ,Transforming Growth Factor beta1 ,Mice ,Rare Diseases ,Stem Cell Research - Nonembryonic - Human ,Animals ,Humans ,2.1 Biological and endogenous factors ,Aetiology ,Lung ,Cultured ,Epithelial Cells ,Cell Differentiation ,Cell Biology ,Fibroblasts ,Biological Sciences ,Stem Cell Research ,Idiopathic Pulmonary Fibrosis ,Pulmonary Alveoli ,Epidermal Cells ,Alveolar Epithelial Cells ,Bone Morphogenetic Proteins ,Cell Transdifferentiation ,Respiratory ,Inbred NOD ,Keratin-5 ,Single-Cell Analysis ,Signal Transduction ,Developmental Biology - Abstract
Loss of alveolar type 2 cells (AEC2s) and the ectopic appearance of basal cells in the alveoli characterize severe lung injuries such as idiopathic pulmonary fibrosis (IPF). Here we demonstrate that human alveolar type 2 cells (hAEC2s), unlike murine AEC2s, transdifferentiate into basal cells in response to fibrotic signalling in the lung mesenchyme, in vitro and in vivo. Single-cell analysis of normal hAEC2s and mesenchymal cells in organoid co-cultures revealed the emergence of pathologic fibroblasts and basaloid cells previously described in IPF. Transforming growth factor-β1 and anti-bone morphogenic protein signalling in the organoids promoted transdifferentiation. Trajectory and histologic analyses of both hAEC2-derived organoids and IPF epithelium indicated that hAEC2s transdifferentiate into basal cells through alveolar-basal intermediates that accumulate in proximity to pathologic CTHRC1hi/TGFB1hi fibroblasts. Our study indicates that hAEC2 loss and expansion of alveolar metaplastic basal cells in severe human lung injuries are causally connected through an hAEC2-basal cell lineage trajectory driven by aberrant mesenchyme.
- Published
- 2022
7. Nuclear IL-33 as a growth and survival agent within basal cells
- Author
-
Harold A. Chapman
- Subjects
medicine.medical_treatment ,Immunology ,Cell ,Biology ,Medical and Health Sciences ,Mice ,Basal (phylogenetics) ,Immune system ,Hypersensitivity ,medicine ,Animals ,Alarmins ,2.1 Biological and endogenous factors ,Aetiology ,Lung ,Cell damage ,Barrier function ,General Medicine ,Interleukin-33 ,medicine.disease ,Interleukin-1 Receptor-Like 1 Protein ,Cell biology ,Interleukin 33 ,medicine.anatomical_structure ,Cytokine ,Cytokines ,Respiratory epithelium ,Research Article - Abstract
Epithelial cells are charged with protection at barrier sites, but whether this normally beneficial response might sometimes become dysfunctional still needs definition. Here, we recognized a pattern of imbalance marked by basal epithelial cell growth and differentiation that replaced normal airspaces in a mouse model of progressive postviral lung disease due to the Sendai virus. Single-cell and lineage-tracing technologies identified a distinct subset of basal epithelial stem cells (basal ESCs) that extended into gas-exchange tissue to form long-term bronchiolar-alveolar remodeling regions. Moreover, this cell subset was selectively expanded by crossing a cell-growth and survival checkpoint linked to the nuclear-localized alarmin IL-33 that was independent of IL-33 receptor signaling and instead connected to autocrine chromatin accessibility. This mechanism creates an activated stem-progenitor cell lineage with potential for physiological or pathological function. Thus, conditional loss of Il33 gene function in basal epithelial cells disrupted the homeostasis of the epithelial barrier at skin and gut sites but also markedly attenuated postviral disease in the lung based on the downregulation of remodeling and inflammation. Thus, we define a basal ESC strategy to deploy innate immune machinery that appears to overshoot the primordial goal of self-defense. Our findings reveal new targets to stratify and correct chronic and often deadly postviral disease.
- Published
- 2021
- Full Text
- View/download PDF
8. IRE1α drives lung epithelial progenitor dysfunction to establish a niche for pulmonary fibrosis
- Author
-
Alexis Brumwell, Harold A. Chapman, Vincent C. Auyeung, Feroz R. Papa, Maike Thamsen, Michael S. Downey, Dean Sheppard, Bradley J. Backes, Jaymin J. Kathiriya, and Talia A. Wenger
- Subjects
education.field_of_study ,Lung ,Population ,Biology ,medicine.disease ,Bleomycin ,Idiopathic pulmonary fibrosis ,chemistry.chemical_compound ,medicine.anatomical_structure ,chemistry ,Fibrosis ,Pulmonary fibrosis ,medicine ,Unfolded protein response ,Cancer research ,Progenitor cell ,education - Abstract
Idiopathic pulmonary fibrosis (IPF) is a disease of progressive interstitial fibrosis, which leads to severe debilitation, respiratory failure, and death. In IPF, environmental exposures interact with genetic risk factors to engender critical patho-etiological events in lung epithelial cells, including endoplasmic reticulum (ER) stress and TGFβ signaling, but the interactions between these disparate pathways are not well understood. We previously showed that kinase inhibitors of the IRE1α bifunctional kinase/RNase—a central mediator of the unfolded protein response (UPR) to ER stress—protected mice from bleomycin-induced pulmonary fibrosis. Here we show that a nanomolar-potent, mono-selective kinase inhibitor of IRE1α (KIRA8) decreases ER-stress induced TGFβ signaling and the senescence-associated secretory phenotype (SASP) in the lung epithelium after bleomycin exposure. A recently-described subset of “damage-associated transient progenitors” (DATPs) display IRE1α-regulated pathological gene signatures that are quelled by KIRA8, in vivo. After injury, these cells uniquely express integrin αvβ6, a key activator of TGFβ in pulmonary fibrosis. KIRA8 inhibition of IRE1α decreases both DATP number and Itgb6 expression in remaining cells, with a decrease in local collagen accumulation. Single-cell RNA sequencing from IPF lungs revealed an analogous Itgb6+ cell population that may also be regulated by IRE1α. These findings suggest that lung epithelial progenitor cells sit at the center of the fibrotic niche, and IRE1α signaling locks them into a dysfunctional state that establishes and perpetuates pathological fibrosis.
- Published
- 2021
- Full Text
- View/download PDF
9. Nfkbid Overexpression in Nonobese Diabetic Mice Elicits Complete Type 1 Diabetes Resistance in Part Associated with Enhanced Thymic Deletion of Pathogenic CD8 T Cells and Increased Numbers and Activity of Regulatory T Cells
- Author
-
Jennifer R. Dwyer, Jeremy J. Racine, Harold D. Chapman, Anna Quinlan, Maximiliano Presa, Grace A. Stafford, Ingo Schmitz, and David V. Serreze
- Subjects
Mice ,Diabetes Mellitus, Type 1 ,Mice, Inbred NOD ,Immunology ,Immunology and Allergy ,Animals ,Humans ,Mice, Transgenic ,CD8-Positive T-Lymphocytes ,T-Lymphocytes, Regulatory ,Diabetes Mellitus, Experimental - Abstract
Type 1 diabetes (T1D) in both humans and NOD mice is caused by T cell–mediated autoimmune destruction of pancreatic β cells. Increased frequency or activity of autoreactive T cells and failures of regulatory T cells (Tregs) to control these pathogenic effectors have both been implicated in T1D etiology. Due to the expression of MHC class I molecules on β cells, CD8 T cells represent the ultimate effector population mediating T1D. Developing autoreactive CD8 T cells normally undergo extensive thymic negative selection, but this process is impaired in NOD mice and also likely T1D patients. Previous studies identified an allelic variant of Nfkbid, a NF-κB signal modulator, as a gene strongly contributing to defective thymic deletion of autoreactive CD8 T cells in NOD mice. These previous studies found ablation of Nfkbid in NOD mice using the clustered regularly interspaced short palindromic repeats system resulted in greater thymic deletion of pathogenic CD8 AI4 and NY8.3 TCR transgenic T cells but an unexpected acceleration of T1D onset. This acceleration was associated with reductions in the frequency of peripheral Tregs. In this article, we report transgenic overexpression of Nfkbid in NOD mice also paradoxically results in enhanced thymic deletion of autoreactive CD8 AI4 T cells. However, transgenic elevation of Nfkbid expression also increased the frequency and functional capacity of peripheral Tregs, in part contributing to the induction of complete T1D resistance. Thus, future identification of a pharmaceutical means to enhance Nfkbid expression might ultimately provide an effective T1D intervention approach.
- Published
- 2021
10. Reversal of TGFβ1-Driven Profibrotic State in Patients with Pulmonary Fibrosis
- Author
-
Angela L. Linderholm, Justin M. Oldham, Genevieve Montas, Claude Jourdan Le Saux, Nancy K. Hills, Scott M. Turner, Kirk D. Jones, Paul J. Wolters, Prerna Kotak, Jeffrey A. Golden, B. Trinh, Darren Leong, Ying Wei, Martin Decaris, Harold A. Chapman, Jin-Woo Song, and Elena Foster
- Subjects
medicine.medical_specialty ,Biopsy ,Pulmonary Fibrosis ,Lung biopsy ,030204 cardiovascular system & hematology ,Epigallocatechin gallate ,complex mixtures ,Gastroenterology ,Antioxidants ,Catechin ,Collagen Type I ,Article ,Transforming Growth Factor beta1 ,03 medical and health sciences ,chemistry.chemical_compound ,0302 clinical medicine ,Internal medicine ,Pulmonary fibrosis ,Humans ,Medicine ,heterocyclic compounds ,In patient ,030212 general & internal medicine ,Lung ,medicine.diagnostic_test ,business.industry ,Extramural ,food and beverages ,General Medicine ,respiratory system ,medicine.disease ,medicine.anatomical_structure ,chemistry ,Snail Family Transcription Factors ,sense organs ,business ,Biomarkers - Abstract
EGCG and Pulmonary Fibrosis Nine patients who were scheduled to undergo lung biopsy for pulmonary fibrosis were given epigallocatechin gallate (EGCG) for 2 weeks. Levels of fibrotic markers in lung...
- Published
- 2020
- Full Text
- View/download PDF
11. Yap/Taz regulate alveolar regeneration and resolution of lung inflammation
- Author
-
Ying Tian, Peggy Zhang, Marla R. Wolfson, Harold A. Chapman, Beata Kosmider, Edward E. Morrisey, Lauren Tragesser, Yan Wang, Daniela Liccardo, Walter J. Koch, Ryan LaCanna, Tongtong Cao, and Hao Shen
- Subjects
Male ,0301 basic medicine ,Pulmonology ,Cell Cycle Proteins ,Stem cells ,Inbred C57BL ,Regenerative Medicine ,Medical and Health Sciences ,Epithelium ,Transgenic ,Mice ,0302 clinical medicine ,Bacterial infections ,2.1 Biological and endogenous factors ,Aetiology ,Lung ,Adult stem cells ,Stem Cells ,NF-kappa B ,Adaptor Proteins ,Cell Differentiation ,General Medicine ,respiratory system ,Pulmonary Surfactant-Associated Protein C ,Cell biology ,Streptococcus pneumoniae ,Infectious Diseases ,medicine.anatomical_structure ,030220 oncology & carcinogenesis ,Pneumonia & Influenza ,Respiratory ,Pneumococcal ,Stem Cell Research - Nonembryonic - Non-Human ,medicine.symptom ,Stem cell ,Infection ,Research Article ,Signal Transduction ,Biotechnology ,Alveolar Epithelium ,Immunology ,Mice, Transgenic ,Inflammation ,03 medical and health sciences ,Rare Diseases ,Downregulation and upregulation ,medicine ,Animals ,Humans ,Regeneration ,Adaptor Proteins, Signal Transducing ,Cell Proliferation ,Cell Nucleus ,Hippo signaling pathway ,business.industry ,Regeneration (biology) ,Signal Transducing ,Epithelial Cells ,YAP-Signaling Proteins ,Pneumonia ,Pneumonia, Pneumococcal ,Stem Cell Research ,medicine.disease ,respiratory tract diseases ,Mice, Inbred C57BL ,HEK293 Cells ,030104 developmental biology ,Alveolar Epithelial Cells ,Trans-Activators ,business - Abstract
Alveolar epithelium plays a pivotal role in protecting the lungs from inhaled infectious agents. Therefore, the regenerative capacity of the alveolar epithelium is critical for recovery from these insults in order to rebuild the epithelial barrier and restore pulmonary functions. Here, we show that sublethal infection of mice with Streptococcus pneumoniae, the most common pathogen of community-acquired pneumonia, led to exclusive damage in lung alveoli, followed by alveolar epithelial regeneration and resolution of lung inflammation. We show that surfactant protein C–expressing (SPC-expressing) alveolar epithelial type II cells (AECIIs) underwent proliferation and differentiation after infection, which contributed to the newly formed alveolar epithelium. This increase in AECII activities was correlated with increased nuclear expression of Yap and Taz, the mediators of the Hippo pathway. Mice that lacked Yap/Taz in AECIIs exhibited prolonged inflammatory responses in the lung and were delayed in alveolar epithelial regeneration during bacterial pneumonia. This impaired alveolar epithelial regeneration was paralleled by a failure to upregulate IκBa, the molecule that terminates NF-κB–mediated inflammatory responses. These results demonstrate that signals governing resolution of lung inflammation were altered in Yap/Taz mutant mice, which prevented the development of a proper regenerative niche, delaying repair and regeneration of alveolar epithelium during bacterial pneumonia.
- Published
- 2019
- Full Text
- View/download PDF
12. Human alveolar type 2 epithelium transdifferentiates into metaplastic KRT5
- Author
-
Jaymin J, Kathiriya, Chaoqun, Wang, Minqi, Zhou, Alexis, Brumwell, Monica, Cassandras, Claude Jourdan, Le Saux, Max, Cohen, Kostantinos-Dionysios, Alysandratos, Bruce, Wang, Paul, Wolters, Michael, Matthay, Darrell N, Kotton, Harold A, Chapman, and Tien, Peng
- Subjects
Cell Differentiation ,Epithelial Cells ,Mice, Transgenic ,Mice, SCID ,Respiratory Mucosa ,Fibroblasts ,Idiopathic Pulmonary Fibrosis ,Mesoderm ,Mice, Inbred C57BL ,Pulmonary Alveoli ,Transforming Growth Factor beta1 ,Mice ,Epidermal Cells ,Mice, Inbred NOD ,Alveolar Epithelial Cells ,Bone Morphogenetic Proteins ,Cell Transdifferentiation ,Animals ,Humans ,Keratin-5 ,Single-Cell Analysis ,Cells, Cultured ,Signal Transduction - Abstract
Loss of alveolar type 2 cells (AEC2s) and the ectopic appearance of basal cells in the alveoli characterize severe lung injuries such as idiopathic pulmonary fibrosis (IPF). Here we demonstrate that human alveolar type 2 cells (hAEC2s), unlike murine AEC2s, transdifferentiate into basal cells in response to fibrotic signalling in the lung mesenchyme, in vitro and in vivo. Single-cell analysis of normal hAEC2s and mesenchymal cells in organoid co-cultures revealed the emergence of pathologic fibroblasts and basaloid cells previously described in IPF. Transforming growth factor-β1 and anti-bone morphogenic protein signalling in the organoids promoted transdifferentiation. Trajectory and histologic analyses of both hAEC2-derived organoids and IPF epithelium indicated that hAEC2s transdifferentiate into basal cells through alveolar-basal intermediates that accumulate in proximity to pathologic CTHRC1
- Published
- 2021
13. Human distal airways contain a multipotent secretory cell that can regenerate alveoli
- Author
-
Maria C. Basil, Fabian L. Cardenas-Diaz, Jaymin J. Kathiriya, Michael P. Morley, Justine Carl, Alexis N. Brumwell, Jeremy Katzen, Katherine J. Slovik, Apoorva Babu, Su Zhou, Madison M. Kremp, Katherine B. McCauley, Shanru Li, Joseph D. Planer, Shah S. Hussain, Xiaoming Liu, Rebecca Windmueller, Yun Ying, Kathleen M. Stewart, Michelle Oyster, Jason D. Christie, Joshua M. Diamond, John F. Engelhardt, Edward Cantu, Steven M. Rowe, Darrell N. Kotton, Harold A. Chapman, and Edward E. Morrisey
- Subjects
Chronic Obstructive ,Multidisciplinary ,General Science & Technology ,Multipotent Stem Cells ,Ferrets ,Stem Cell Research - Embryonic - Non-Human ,Stem Cell Research ,Article ,Pulmonary Disease ,Pulmonary Alveoli ,Mice ,Pulmonary Disease, Chronic Obstructive ,Respiratory ,2.1 Biological and endogenous factors ,Animals ,Humans ,Cell Lineage ,Aetiology ,Bronchioles ,Lung - Abstract
The human lung differs substantially from its mouse counterpart, resulting in a distinct distal airway architecture affected by disease pathology in chronic obstructive pulmonary disease. In humans, the distal branches of the airway interweave with the alveolar gas-exchange niche, forming an anatomical structure known as the respiratory bronchioles. Owing to the lack of a counterpartin mouse, the cellular and molecular mechanisms that governrespiratory bronchioles in the human lung remain uncharacterized. Here we show that human respiratory bronchioles contain a unique secretory cell population that is distinct from cells in larger proximal airways. Organoid modelling reveals that these respiratory airway secretory (RAS) cells act as unidirectional progenitors for alveolar type 2 cells, which are essential for maintaining and regenerating the alveolar niche. RAS cell lineage differentiation into alveolar type 2 cells is regulated by Notch and Wnt signalling. In chronic obstructive pulmonary disease, RAS cells are altered transcriptionally, corresponding to abnormal alveolar type 2 cell states, which are associated with smoking exposure in both humans and ferrets. These data identify a distinct progenitor in a region of the human lung that is not found in mouse that has a critical role in maintaining the gas-exchange compartment and is altered in chronic lung disease.
- Published
- 2021
14. Collagen promotes anti-PD-1/PD-L1 resistance in cancer through LAIR1-dependent CD8+ T cell exhaustion
- Author
-
Carmen Behrens, Ying Wei, Bertha Leticia Rodriguez, Jing Wang, Lixia Diao, Edwin Roger Parra Cuentes, Luisa M. Solis Soto, Gabriela Raso, Harold A. Chapman, Ignacio I. Wistuba, David H. Peng, Don L. Gibbons, Jonathan M. Kurie, Lauren Averett Byers, Mitsuo Yamauchi, and Limo Chen
- Subjects
0301 basic medicine ,Male ,Lung Neoplasms ,medicine.medical_treatment ,Programmed Cell Death 1 Receptor ,Drug Resistance ,General Physics and Astronomy ,Datasets as Topic ,Cancer immunotherapy ,CD8-Positive T-Lymphocytes ,Transgenic ,B7-H1 Antigen ,Extracellular matrix ,Mice ,Non-Receptor Type 6 ,0302 clinical medicine ,Immunologic ,Receptors ,Cytotoxic T cell ,2.1 Biological and endogenous factors ,RNA-Seq ,Aetiology ,lcsh:Science ,Lung ,Hepatitis A Virus Cellular Receptor 2 ,Cancer ,Multidisciplinary ,Tumor ,biology ,LOXL2 ,Chemistry ,Lung Cancer ,Extracellular Matrix ,medicine.anatomical_structure ,Immunological ,5.1 Pharmaceuticals ,030220 oncology & carcinogenesis ,Gene Knockdown Techniques ,Tumour immunology ,Female ,Collagen ,Amino Acid Oxidoreductases ,Lung cancer ,Development of treatments and therapeutic interventions ,Cancer microenvironment ,Science ,T cell ,Antineoplastic Agents ,Adenocarcinoma of Lung ,Article ,General Biochemistry, Genetics and Molecular Biology ,Cell Line ,Proto-Oncogene Proteins p21(ras) ,03 medical and health sciences ,Immune system ,Clinical Research ,PD-L1 ,medicine ,Animals ,Humans ,Lewis Lung ,Animal ,Carcinoma ,General Chemistry ,Immunotherapy ,030104 developmental biology ,HEK293 Cells ,Good Health and Well Being ,Disease Models ,Cancer research ,biology.protein ,Neoplasm ,lcsh:Q ,Protein Tyrosine Phosphatase ,CD8 ,Biomarkers - Abstract
Tumor extracellular matrix has been associated with drug resistance and immune suppression. Here, proteomic and RNA profiling reveal increased collagen levels in lung tumors resistant to PD-1/PD-L1 blockade. Additionally, elevated collagen correlates with decreased total CD8+ T cells and increased exhausted CD8+ T cell subpopulations in murine and human lung tumors. Collagen-induced T cell exhaustion occurs through the receptor LAIR1, which is upregulated following CD18 interaction with collagen, and induces T cell exhaustion through SHP-1. Reduction in tumor collagen deposition through LOXL2 suppression increases T cell infiltration, diminishes exhausted T cells, and abrogates resistance to anti-PD-L1. Abrogating LAIR1 immunosuppression through LAIR2 overexpression or SHP-1 inhibition sensitizes resistant lung tumors to anti-PD-1. Clinically, increased collagen, LAIR1, and TIM-3 expression in melanoma patients treated with PD-1 blockade predict poorer survival and response. Our study identifies collagen and LAIR1 as potential markers for immunotherapy resistance and validates multiple promising therapeutic combinations., Tumor extracellular matrix has been associated with cancer progression, therapy resistance and immune suppression. Here, the authors show that collagen generates resistance to PD-1/PD-L1 immunotherapy by upregulating LAIR1 expression and downstream signaling, leading to increased CD8+ T cell exhaustion.
- Published
- 2020
15. Blocking LOXL2 and TGFβ1 signalling induces collagen I turnover in precision-cut lung slices derived from patients with idiopathic pulmonary fibrosis
- Author
-
Tsung-Che Ho, Xiaopeng Li, Harold A. Chapman, Wenting Dong, Max L Cohen, Julia R. Jackson, Ying Wei, Paul J. Wolters, Claude Jourdan Le Saux, Julia Klesney-Tait, and Alexis N. Brumwell
- Subjects
Pulmonary and Respiratory Medicine ,Immunoblotting ,Epigallocatechin gallate ,Collagen Type I ,Article ,Transforming Growth Factor beta1 ,03 medical and health sciences ,Idiopathic pulmonary fibrosis ,chemistry.chemical_compound ,0302 clinical medicine ,Fibrosis ,Pulmonary fibrosis ,medicine ,Humans ,Receptor ,Lung ,Cells, Cultured ,030304 developmental biology ,0303 health sciences ,LOXL2 ,business.industry ,medicine.disease ,Idiopathic Pulmonary Fibrosis ,Transplantation ,medicine.anatomical_structure ,030228 respiratory system ,chemistry ,Cancer research ,Amino Acid Oxidoreductases ,business ,Signal Transduction - Abstract
We recently identified epigallocatechin gallate (EGCG), a trihydroxyphenolic compound, as a dual inhibitor of lysyl oxidase-like2 and transforming growth factor-β1 (TGFβ1) receptor kinase that when given orally to patients with idiopathic pulmonary fibrosis (IPF) reversed profibrotic biomarkers in their diagnostic biopsies. Here, we extend these findings to advanced pulmonary fibrosis using cultured precision-cut lung slices from explants of patients with IPF undergoing transplantation. During these experiments, we were surprised to discover that not only did EGCG attenuate TGFβ1 signalling and new collagen accumulation but also activated matrix metalloproteinase-dependent collagen I turnover, raising the possibility of slow fibrosis resolution with continued treatment.
- Published
- 2020
16. Human alveolar Type 2 epithelium transdifferentiates into metaplastic KRT5+ basal cells during alveolar repair
- Author
-
Michael A. Matthay, Monica Cassandras, Chaoqun Wang, Harold A. Chapman, Alexis N. Brumwell, Jaymin J. Kathiriya, Paul J. Wolters, Claude Le Saux, and Tien Peng
- Subjects
Basal (phylogenetics) ,medicine.anatomical_structure ,Immunophenotyping ,Mesenchyme ,Transdifferentiation ,medicine ,Organoid ,Stem cell ,Progenitor cell ,Biology ,Epithelium ,Cell biology - Abstract
SUMMARYUnderstanding differential lineage potential of orthologous stem cells across species can shed light on human disease. Here, utilizing 3D organoids, single cell RNAseq, and xenotransplants, we demonstrate that human alveolar type 2 cells (hAEC2s), unlike murine AEC2s, are multipotent and able to transdifferentiate into KRT5+ basal cells when co-cultured with primary fibroblasts in 3D organoids. Trajectory analyses and immunophenotyping of epithelial progenitors in idiopathic pulmonary fibrosis (IPF) indicate that hAEC2s transdifferentiate into metaplastic basal cells through alveolar-basal intermediate (ABI) cells that we also identify in hAEC2-derived organoids. Modulating hAEC2-intrinsic and niche factors dysregulated in IPF can attenuate metaplastic basal cell transdifferentiation and preserve hAEC2 identity. Finally, hAEC2s transplanted into fibrotic immune-deficient murine lungs engraft as either hAEC2s or differentiated KRT5+ basal cells. Our study indicates that hAEC2s-loss and expansion of alveolar metaplastic basal cells in IPF are causally connected, which would not have been revealed utilizing murine AEC2s as a model.HighlightsHuman AEC2s transdifferentiate into KRT5+ basal cells when accompanied by primary adult human lung mesenchyme in 3D organoid culture.Alterations of hAEC2-intrinsic and niche factors dysregulated in IPF can modify metaplastic hAEC2 transdifferentiation.hAEC2s engraft into fibrotic lungs of immune-deficient mice and transdifferentiate into metaplastic basal cells.Transcriptional trajectory analysis suggests that hAEC2s in IPF gives rise to metaplastic basal cells via alveolar-basal intermediate cells.
- Published
- 2020
- Full Text
- View/download PDF
17. Human CD8+ T-cells Require Glycolysis to Elicit Effector Function
- Author
-
Thomas E. Angelini, Matthew E. Merritt, Scott E. Stimpson, Brittney N. Newby, Ram B. Khattri, Clayton E. Mathews, Harold D. Chapman, David V. Serreze, and Jiang Chen
- Subjects
0303 health sciences ,Chemistry ,Effector ,Nod ,Metabolism ,3. Good health ,Cell biology ,03 medical and health sciences ,CTL ,0302 clinical medicine ,Immune system ,Lytic cycle ,Cytotoxic T cell ,Glycolysis ,030217 neurology & neurosurgery ,030304 developmental biology - Abstract
Targeting human T-cell metabolism for modulating immune function requires an understanding of macronutrient utilization. Using metabolic inhibition during activation of human naïve CD8+ T-cells, we demonstrate blocking glycolysis or mitochondrial respiration prevents T-cell proliferation. However, after activation and differentiation, the metabolic program changes. Inhibition of glycolysis abolished cytotoxic T-lymphocyte (CTL) activity, whereas mitochondrial inhibition had no effect on CTL lytic function. Studies with uniformly labeled 13C-glucose confirmed CTL convert the majority of glucose to lactate. The role of glycolysis in CTL function was assessed using NOD models of Type 1 diabetes (T1D). Treatment of NOD models with a glycolysis inhibitor resulted in reduced and delayed T1D incidence and significantly preserved β-cell mass. We conclude glycolysis and mitochondrial ATP production are essential for efficient T-cell activation, but only glycolysis is essential for CTL lytic function. These data suggest targeting glycolysis in CTLs is a promising pathway to prevent T-cell-mediated autoimmunity.
- Published
- 2020
- Full Text
- View/download PDF
18. T Cells from NOD
- Author
-
Jeremy J, Racine, Harold D, Chapman, Rosalinda, Doty, Brynn M, Cairns, Timothy J, Hines, Abigail L D, Tadenev, Laura C, Anderson, Torrian, Green, Meaghan E, Dyer, Janine M, Wotton, Zoë, Bichler, Jacqueline K, White, Rachel, Ettinger, Robert W, Burgess, and David V, Serreze
- Subjects
CD4-Positive T-Lymphocytes ,Mice ,Diabetes Mellitus, Type 1 ,Mice, Inbred NOD ,Animals ,Mice, Transgenic ,Mice, SCID ,CD8-Positive T-Lymphocytes ,Nerve Tissue ,Neuritis, Autoimmune, Experimental ,Pancreas ,Article ,Diabetes Mellitus, Experimental - Abstract
It has become increasingly appreciated that autoimmune responses against neuronal components play an important role in type 1 diabetes (T1D(4)) pathogenesis. In fact, a large proportion of islet-infiltrating B-lymphocytes in the NOD mouse model of T1D produce antibodies directed against the neuronal type III intermediate filament protein, peripherin. NOD-PerIg mice are a previously developed BCR-transgenic model in which virtually all B-lymphocytes express the heavy and light chain immunoglobulin molecules from the intra-islet derived anti-peripherin reactive hybridoma H280. NOD-PerIg mice have accelerated T1D development and PerIg B-lymphocytes actively proliferate within islets and expand cognitively interactive pathogenic T-cells from a pool of naïve precursors. We now report adoptively transferred T-cells or whole splenocytes from NOD-PerIg mice expectedly induce T1D in NOD.scid recipients, but depending on the kinetics of disease development, can also elicit a peripheral neuritis (with secondary myositis). This neuritis was predominantly composed of CD4(+) and CD8(+) T-cells. Antibody depletion studies showed neuritis still developed in the absence of NOD-PerIg CD8(+) T-cells, but required CD4(+) T-cells. Surprisingly, sciatic nerve-infiltrating CD4(+) cells had an expansion of IFN-γ(−) and TNF-α(−) double negative cells compared to those within both islets and spleen. Nerve and islet infiltrating CD4(+) T-cells also differed by expression patterns of CD95, PD-1 and Tim-3. Further studies found transitory early B-lymphocyte depletion delayed T1D onset in a portion of NOD-PerIg mice allowing them to survive long enough to develop neuritis outside of the transfer setting. Together, this study presents a new model of peripherin-reactive B-lymphocyte dependent autoimmune neuritis.
- Published
- 2020
19. Distinct airway epithelial stem cells hide among club cells but mobilize to promote alveolar regeneration
- Author
-
Alexis N. Brumwell, Jaymin J. Kathiriya, Julia R. Jackson, Xiaodan Tang, and Harold A. Chapman
- Subjects
bleomycin injury ,lung epithelial stem cells ,Lung injury ,Biology ,single cell transcriptomics ,Regenerative Medicine ,Medical and Health Sciences ,Article ,Alveolar cells ,03 medical and health sciences ,0302 clinical medicine ,Rare Diseases ,Stem Cell Research - Nonembryonic - Human ,Genetics ,medicine ,Humans ,Progenitor cell ,Bronchioles ,Lung ,030304 developmental biology ,0303 health sciences ,Regeneration (biology) ,MHC(high) airway progenitors ,Stem Cells ,dedifferentiation ,Cell Differentiation ,Epithelial Cells ,Cell Biology ,Lung Injury ,Biological Sciences ,respiratory system ,Stem Cell Research ,Epithelium ,Cell biology ,respiratory tract diseases ,Transplantation ,alveolar injury and regeneration ,medicine.anatomical_structure ,Alveolar Epithelial Cells ,Respiratory ,Molecular Medicine ,Stem Cell Research - Nonembryonic - Non-Human ,oxygenation ,Stem cell ,030217 neurology & neurosurgery ,transplantation ,Developmental Biology - Abstract
Lung injury activates specialized adult epithelial progenitors to regenerate the epithelium. Depending on the extent of injury, both remaining alveolar type II cells (AEC2s) and distal airway stem/progenitors mobilize to cover denuded alveoli and restore normal barriers. The major source of airway stem/progenitors other than basal-like cells remains uncertain. Here, we define a distinct subpopulation (∼5%) of club-like lineage-negative epithelial progenitors (LNEPs) marked by high H2-K1 expression critical for alveolar repair. Quiescent H2-K1high cells account for virtually all invitro regenerative activity of airway lineages. After bleomycin injury, H2-K1 cells expand and differentiate invivo to alveolar lineages. However,injured H2-K1 cells eventually develop impaired self-renewal with features of senescence, limiting complete repair. Normal H2-K1high cells transplanted into injured lungs differentiate into alveolar cells and rescue lung function. These findings indicate that small subpopulations of specialized stem/progenitors are required for effective lung regeneration and are a potential therapeutic adjunct after major lung injury.
- Published
- 2020
20. Contextual Cues from Cancer Cells Govern CAF Heterogeneity
- Author
-
Jing Wang, Russell William, Jordan S. Miller, Daniel W. Sazer, Chad J. Creighton, Xin Liu, Michael weiger, Hou-Fu Guo, Barbara Mino, Kuanwei Sheng, Neus Bota-Rabassedas, Gregory D. Longmore, Joshua J.A. Firestone, Maria Gabriela Raso, Pamela Villalobos, B. Leticia Rodriguez, Jaime Rodriguez-Canales, Yuanxin Xi, Ignacio I. Wistuba, Yichi Niu, Priyam Banerjee, Jonathan M. Kurie, Edwin Roger Parra, Wenjian Cao, Don L. Gibbons, Xiaochao Tan, Jiayi Luo, Luisa S Solis, Jiang Yu, Jacob L. Albritton, Chenghang Zong, and Harold A. Chapman
- Subjects
Tumor microenvironment ,Stromal cell ,Single cell sequencing ,Cancer cell ,medicine ,Cancer-Associated Fibroblasts ,Adenocarcinoma ,Biology ,medicine.disease ,Reprogramming ,Phenotype ,Cell biology - Abstract
Cancer cells function as primary architects of the tumor microenvironment. Yet, the molecular features of cancer cells that govern stromal cell phenotypes remain unclear. Here, we show that cancer-associated fibroblasts (CAFs) are distinguishable on the basis of gene expression signatures they acquire in co-culture with epithelial- or mesenchymal-like lung adenocarcinoma (LUAD) cells. High expression of the EMT activator ZEB1 endows LUAD cells with the capacity to activate a soluble factor exchange that leads to CAF reprogramming, to generate CAF-led invasive projections in multicellular aggregates, and to respond to pro-metastatic signals from CAFs in mice. Thus, ZEB1-expressing LUAD cells are positioned at the apex of a signaling hierarchy in the tumor microenvironment.
- Published
- 2020
- Full Text
- View/download PDF
21. A MusD retrotransposon insertion in the mouse Slc6a5 gene causes alterations in neuromuscular junction maturation and behavioral phenotypes.
- Author
-
Laurent P Bogdanik, Harold D Chapman, Kathy E Miers, David V Serreze, and Robert W Burgess
- Subjects
Medicine ,Science - Abstract
Glycine is the major inhibitory neurotransmitter in the spinal cord and some brain regions. The presynaptic glycine transporter, GlyT2, is required for sustained glycinergic transmission through presynaptic reuptake and recycling of glycine. Mutations in SLC6A5, encoding GlyT2, cause hereditary hyperekplexia in humans, and similar phenotypes in knock-out mice, and variants are associated with schizophrenia. We identified a spontaneous mutation in mouse Slc6a5, caused by a MusD retrotransposon insertion. The GlyT2 protein is undetectable in homozygous mutants, indicating a null allele. Homozygous mutant mice are normal at birth, but develop handling-induced spasms at five days of age, and only survive for two weeks, but allow the study of early activity-regulated developmental processes. At the neuromuscular junction, synapse elimination and the switch from embryonic to adult acetylcholine receptor subunits are hastened, consistent with a presumed increase in motor neuron activity, and transcription of acetylcholine receptors is elevated. Heterozygous mice, which show no reduction in lifespan but nonetheless have reduced levels of GlyT2, have a normal thermal sensitivity with the hot-plate test, but differences in repetitive grooming and decreased sleep time with home-cage monitoring. Open-field and elevated plus-maze tests did not detect anxiety-like behaviors; however, the latter showed a hyperactivity phenotype. Importantly, grooming and hyperactivity are observed in mouse schizophrenia models. Thus, mutations in Slc6a5 show changes in neuromuscular junction development as homozygotes, and behavioral phenotypes as heterozygotes, indicating their usefulness for studies related to glycinergic dysfunction.
- Published
- 2012
- Full Text
- View/download PDF
22. Extracellular matrix in lung development, homeostasis and disease
- Author
-
Alexandra Naba, Jenna L. Balestrini, Peter B. Bitterman, Luis A. Ortiz, Kirk C. Hansen, Daniel J. Tschumperlin, Yong Zhou, Farrah Kheradmand, Namasivayam Ambalavanan, Laura E. Niklason, Kamran Atabai, William C. Parks, Adam J. Engler, James S. Hagood, Eric S. White, Richard A. Corley, Enid Neptune, Jeffrey C. Horowitz, Harold A. Chapman, Victor J. Thannickal, Qing S. Lin, and Bi-Sen Ding
- Subjects
Lung Diseases ,0301 basic medicine ,Biochemistry & Molecular Biology ,1.1 Normal biological development and functioning ,Disease ,Biology ,Regenerative Medicine ,Article ,Pathogenesis ,Extracellular matrix ,03 medical and health sciences ,Underpinning research ,medicine ,Humans ,Homeostasis ,Lung ,Molecular Biology ,Cancer ,Uncategorized ,Extracellular Matrix Proteins ,Regeneration (biology) ,Lung Cancer ,Biological Sciences ,respiratory system ,Stem Cell Research ,Phenotype ,Extracellular Matrix ,Biomechanical Phenomena ,respiratory tract diseases ,Cell biology ,030104 developmental biology ,medicine.anatomical_structure ,Respiratory ,Stem Cell Research - Nonembryonic - Non-Human ,Function (biology) - Abstract
The lung's unique extracellular matrix (ECM), while providing structural support for cells, is critical in the regulation of developmental organogenesis, homeostasis and injury-repair responses. The ECM, via biochemical or biomechanical cues, regulates diverse cell functions, fate and phenotype. The composition and function of lung ECM become markedly deranged in pathological tissue remodeling. ECM-based therapeutics and bioengineering approaches represent promising novel strategies for regeneration/repair of the lung and treatment of chronic lung diseases. In this review, we assess the current state of lung ECM biology, including fundamental advances in ECM composition, dynamics, topography, and biomechanics; the role of the ECM in normal and aberrant lung development, adult lung diseases and autoimmunity; and ECM in the regulation of the stem cell niche. We identify opportunities to advance the field of lung ECM biology and provide a set recommendations for research priorities to advance knowledge that would inform novel approaches to the pathogenesis, diagnosis, and treatment of chronic lung diseases.
- Published
- 2018
- Full Text
- View/download PDF
23. Expansion of hedgehog disrupts mesenchymal identity and induces emphysema phenotype
- Author
-
Julie B. Sneddon, Alfred Li, Tien Peng, Nabora S. Reyes de Mochel, Lauren Byrnes, Stephanie A. Christenson, Chaoqun Wang, Alexis N. Brumwell, Yasuyuki Yokosaki, Monica Cassandras, Patty J. Lee, Peiying Shan, Harold A. Chapman, Rebecca Moon, David M. Jablons, and Michael A. Matthay
- Subjects
0301 basic medicine ,Pulmonology ,Knockout ,Mesenchyme ,Immunology ,Population ,Development ,Signal transduction ,Biology ,Medical and Health Sciences ,Mice ,03 medical and health sciences ,0302 clinical medicine ,medicine ,COPD ,2.1 Biological and endogenous factors ,Animals ,Humans ,Hedgehog Proteins ,Aetiology ,Fibroblast ,education ,Lung ,Hedgehog ,Adult stem cells ,Mice, Knockout ,education.field_of_study ,Mesenchymal stem cell ,General Medicine ,Fibroblasts ,respiratory system ,Stem Cell Research ,Hedgehog signaling pathway ,Cell biology ,Pulmonary Alveoli ,030104 developmental biology ,medicine.anatomical_structure ,Pulmonary Emphysema ,Stem Cell Research - Nonembryonic - Non-Human ,Stem cell ,030217 neurology & neurosurgery ,Signal Transduction ,Research Article ,Adult stem cell - Abstract
GWAS have repeatedly mapped susceptibility loci for emphysema to genes that modify hedgehog signaling, but the functional relevance of hedgehog signaling to this morbid disease remains unclear. In the current study, we identified a broad population of mesenchymal cells in the adult murine lung receptive to hedgehog signaling, characterized by higher activation of hedgehog surrounding the proximal airway relative to the distal alveoli. Single-cell RNA-sequencing showed that the hedgehog-receptive mesenchyme is composed of mostly fibroblasts with distinct proximal and distal subsets with discrete identities. Ectopic hedgehog activation in the distal fibroblasts promoted expression of proximal fibroblast markers and loss of distal alveoli and airspace enlargement of over 20% compared with controls. We found that hedgehog suppressed mesenchymal-derived mitogens enriched in distal fibroblasts that regulate alveolar stem cell regeneration and airspace size. Finally, single-cell analysis of the human lung mesenchyme showed that segregated proximal-distal identity with preferential hedgehog activation in the proximal fibroblasts was conserved between mice and humans. In conclusion, we showed that differential hedgehog activation segregates mesenchymal identities of distinct fibroblast subsets and that disruption of fibroblast identity can alter the alveolar stem cell niche, leading to emphysematous changes in the murine lung.
- Published
- 2018
- Full Text
- View/download PDF
24. HLA-B*39:06 Efficiently Mediates Type 1 Diabetes in a Mouse Model Incorporating Reduced Thymic Insulin Expression
- Author
-
David V. Serreze, Teresa P. DiLorenzo, Jeremy J. Racine, Jennifer Schloss, Riyasat Ali, and Harold D. Chapman
- Subjects
0301 basic medicine ,T cell ,medicine.medical_treatment ,Immunology ,Genes, MHC Class I ,Mice, Transgenic ,Thymus Gland ,Human leukocyte antigen ,CD8-Positive T-Lymphocytes ,Major histocompatibility complex ,Article ,Mice ,03 medical and health sciences ,0302 clinical medicine ,Mice, Inbred NOD ,Insulin-Secreting Cells ,HLA-A2 Antigen ,medicine ,Animals ,Insulin ,Immunology and Allergy ,Cytotoxic T cell ,Alleles ,NOD mice ,Type 1 diabetes ,biology ,Pancreatic islets ,medicine.disease ,Disease Models, Animal ,Diabetes Mellitus, Type 1 ,030104 developmental biology ,medicine.anatomical_structure ,HLA-B Antigens ,biology.protein ,030215 immunology - Abstract
Type 1 diabetes (T1D) is characterized by T cell–mediated destruction of the insulin-producing β cells of the pancreatic islets. Among the loci associated with T1D risk, those most predisposing are found in the MHC region. HLA-B*39:06 is the most predisposing class I MHC allele and is associated with an early age of onset. To establish an NOD mouse model for the study of HLA-B*39:06, we expressed it in the absence of murine class I MHC. HLA-B*39:06 was able to mediate the development of CD8 T cells, support lymphocytic infiltration of the islets, and confer T1D susceptibility. Because reduced thymic insulin expression is associated with impaired immunological tolerance to insulin and increased T1D risk in patients, we incorporated this in our model as well, finding that HLA-B*39:06–transgenic NOD mice with reduced thymic insulin expression have an earlier age of disease onset and a higher overall prevalence as compared with littermates with typical thymic insulin expression. This was despite virtually indistinguishable blood insulin levels, T cell subset percentages, and TCR Vβ family usage, confirming that reduced thymic insulin expression does not impact T cell development on a global scale. Rather, it will facilitate the thymic escape of insulin-reactive HLA-B*39:06–restricted T cells, which participate in β cell destruction. We also found that in mice expressing either HLA-B*39:06 or HLA-A*02:01 in the absence of murine class I MHC, HLA transgene identity alters TCR Vβ usage by CD8 T cells, demonstrating that some TCR Vβ families have a preference for particular class I MHC alleles.
- Published
- 2018
- Full Text
- View/download PDF
25. Local lung hypoxia determines epithelial fate decisions during alveolar regeneration
- Author
-
Jeffrey E. Gotts, Dong-Kee Lee, Ying Wei, Ying Xi, Thomas J. Kim, Jianming Xu, Harold A. Chapman, Michael A. Matthay, Victor M. Tan, Andrew E. Vaughan, John M. Shannon, Ian Driver, Julia R. Jackson, and Alexis N. Brumwell
- Subjects
Male ,0301 basic medicine ,Time Factors ,Regenerative Medicine ,Medical and Health Sciences ,Transgenic ,Mice ,Influenza A Virus, H1N1 Subtype ,Cell Movement ,Metaplasia ,Receptors ,Influenza A Virus ,2.1 Biological and endogenous factors ,Aetiology ,Stem Cell Niche ,Hypoxia ,Lung ,Wnt Signaling Pathway ,Cells, Cultured ,Cultured ,Receptors, Notch ,biology ,Stem Cells ,Transdifferentiation ,Wnt signaling pathway ,Biological Sciences ,Squamous metaplasia ,3. Good health ,Cell biology ,Phenotype ,Cell Transdifferentiation ,Respiratory ,Stem Cell Research - Nonembryonic - Non-Human ,Female ,Hypoxia-Inducible Factor 1 ,Single-Cell Analysis ,medicine.symptom ,Human ,Notch ,Beta-catenin ,Genotype ,1.1 Normal biological development and functioning ,Cells ,Mice, Transgenic ,alpha Subunit ,Article ,03 medical and health sciences ,Cytokeratin ,Orthomyxoviridae Infections ,Underpinning research ,Influenza, Human ,medicine ,Regeneration ,Animals ,Humans ,Cell Lineage ,H1N1 Subtype ,Cell Proliferation ,Animal ,Tumor Suppressor Proteins ,Gene Expression Profiling ,SOXB1 Transcription Factors ,Epithelial Cells ,Surfactant protein C ,Cell Biology ,Hypoxia-Inducible Factor 1, alpha Subunit ,Stem Cell Research ,Phosphoproteins ,medicine.disease ,Influenza ,Oxygen ,Pulmonary Alveoli ,Disease Models, Animal ,030104 developmental biology ,Disease Models ,Trans-Activators ,biology.protein ,Keratin-5 ,Transcription Factors ,Developmental Biology - Abstract
After influenza infection, lineage-negative epithelial progenitors (LNEPs) exhibit a binary response to reconstitute epithelial barriers: activating a Notch-dependent ΔNp63/cytokeratin 5 (Krt5) remodelling program or differentiating into alveolar type II cells (AEC2s). Here we show that local lung hypoxia, through hypoxia-inducible factor (HIF1α), drives Notch signallingandKrt5pos basal-like cell expansion. Single-cell transcriptional profiling of human AEC2s from fibrotic lungs revealed a hypoxic subpopulation with activated Notch, suppressed surfactant protein C (SPC), and transdifferentiation toward a Krt5pos basal-likestate. Activated murine Krt5pos LNEPs and diseased human AEC2s upregulate strikingly similar core pathways underlyingmigration and squamous metaplasia. While robust, HIF1α-driven metaplasia is ultimately inferior to AEC2 reconstitution inrestoring normal lung function. HIF1α deletion or enhanced Wnt/β-catenin activity in Sox2pos LNEPs blocks Notch andKrt5activation, instead promoting rapid AEC2 differentiation and migration and improving the quality of alveolar repair.
- Published
- 2017
- Full Text
- View/download PDF
26. Genetic and Small Molecule Disruption of the AID/RAD51 Axis Similarly Protects Nonobese Diabetic Mice from Type 1 Diabetes through Expansion of Regulatory B Lymphocytes
- Author
-
Vivek Philip, David V. Serreze, Clive Wasserfall, William H. Schott, Caroline M. Leeth, Jeremy J. Racine, Qiming Wang, Muneer G. Hasham, Harold D. Chapman, Kevin D. Mills, Jeremy J. Ratiu, Jing Zhu, Jane Branca, Mark A. Atkinson, and Nina M Donghia
- Subjects
0301 basic medicine ,T cell ,Regulatory B cells ,Immunology ,RAD51 ,Somatic hypermutation ,4,4'-Diisothiocyanostilbene-2,2'-Disulfonic Acid ,Biology ,Lymphocyte Activation ,Article ,Diabetes Mellitus, Experimental ,Mice ,03 medical and health sciences ,0302 clinical medicine ,Mice, Inbred NOD ,Cytidine Deaminase ,medicine ,Animals ,Immunology and Allergy ,5'-Nucleotidase ,Autoantibodies ,NOD mice ,B-Lymphocytes, Regulatory ,Nuclear Proteins ,RNA-Binding Proteins ,Cytidine deaminase ,Immunoglobulin Class Switching ,Cell biology ,DNA-Binding Proteins ,Diabetes Mellitus, Type 1 ,030104 developmental biology ,medicine.anatomical_structure ,Immunoglobulin class switching ,Somatic Hypermutation, Immunoglobulin ,CRISPR-Cas Systems ,Carrier Proteins ,Homologous recombination ,030215 immunology - Abstract
B lymphocytes play a key role in type 1 diabetes (T1D) development by serving as a subset of APCs preferentially supporting the expansion of autoreactive pathogenic T cells. As a result of their pathogenic importance, B lymphocyte–targeted therapies have received considerable interest as potential T1D interventions. Unfortunately, the B lymphocyte–directed T1D interventions tested to date failed to halt β cell demise. IgG autoantibodies marking humans at future risk for T1D indicate that B lymphocytes producing them have undergone the affinity-maturation processes of class switch recombination and, possibly, somatic hypermutation. This study found that CRISPR/Cas9-mediated ablation of the activation-induced cytidine deaminase gene required for class switch recombination/somatic hypermutation induction inhibits T1D development in the NOD mouse model. The activation-induced cytidine deaminase protein induces genome-wide DNA breaks that, if not repaired through RAD51-mediated homologous recombination, result in B lymphocyte death. Treatment with the RAD51 inhibitor 4,4′-diisothiocyanatostilbene-2, 2′-disulfonic acid also strongly inhibited T1D development in NOD mice. The genetic and small molecule–targeting approaches expanded CD73+ B lymphocytes that exert regulatory activity suppressing diabetogenic T cell responses. Hence, an initial CRISPR/Cas9-mediated genetic modification approach has identified the AID/RAD51 axis as a target for a potentially clinically translatable pharmacological approach that can block T1D development by converting B lymphocytes to a disease-inhibitory CD73+ regulatory state.
- Published
- 2017
- Full Text
- View/download PDF
27. VEGF Drives the Car toward Better Gas Exchange
- Author
-
Harold A. Chapman and Jaymin J. Kathiriya
- Subjects
Vascular Endothelial Growth Factor A ,Endothelium ,VEGF receptors ,Population ,Medical and Health Sciences ,General Biochemistry, Genetics and Molecular Biology ,Mice ,03 medical and health sciences ,0302 clinical medicine ,Genetics ,medicine ,Animals ,education ,Lung ,Molecular Biology ,book ,030304 developmental biology ,0303 health sciences ,education.field_of_study ,biology ,Developmental cell ,Structural integrity ,Cell Biology ,Biological Sciences ,respiratory system ,Cell biology ,Pulmonary Alveoli ,Endothelial stem cell ,Vascular endothelial growth factor A ,medicine.anatomical_structure ,Lung epithelium ,biology.protein ,book.journal ,Automobiles ,030217 neurology & neurosurgery ,Developmental Biology - Abstract
How lung epithelium and endothelium co-develop to maintain structural integrity of alveoli remains unclear. In this issue of Developmental Cell, Ellis etal. define how epithelial Vegfa directs development of a distinct endothelial cell population that ultimately plays a critical role in ensuring appropriate alveolar septation during alveologenesis.
- Published
- 2020
- Full Text
- View/download PDF
28. Interferon-γ Limits Diabetogenic CD8+ T-Cell Effector Responses in Type 1 Diabetes
- Author
-
Brittney N. Newby, David V. Serreze, Cheng Ye, John P. Driver, Yi-Guang Chen, Clayton E. Mathews, Harold D. Chapman, Jeremy J. Racine, Todd M. Brusko, Deanna J. Lamont, and Caroline M. Leeth
- Subjects
0301 basic medicine ,Adoptive cell transfer ,Endocrinology, Diabetes and Metabolism ,Lymphocyte ,Nod ,Biology ,Proinflammatory cytokine ,03 medical and health sciences ,030104 developmental biology ,medicine.anatomical_structure ,Antigen ,Immunology ,Internal Medicine ,medicine ,Cytotoxic T cell ,STAT4 ,CD8 - Abstract
Type 1 diabetes development in the NOD mouse model is widely reported to be dependent on high-level production by autoreactive CD4+ and CD8+ T cells of interferon-γ (IFN-γ), generally considered a proinflammatory cytokine. However, IFN-γ can also participate in tolerance-induction pathways, indicating it is not solely proinflammatory. This study addresses how IFN-γ can suppress activation of diabetogenic CD8+ T cells. CD8+ T cells transgenically expressing the diabetogenic AI4 T-cell receptor adoptively transferred disease to otherwise unmanipulated NOD.IFN-γnull, but not standard NOD, mice. AI4 T cells only underwent vigorous intrasplenic proliferation in NOD.IFN-γnull recipients. Disease-protective IFN-γ could be derived from any lymphocyte source and suppressed diabetogenic CD8+ T-cell responses both directly and through an intermediary nonlymphoid cell population. Suppression was not dependent on regulatory T cells, but was associated with increased inhibitory STAT1 to STAT4 expression levels in pathogenic AI4 T cells. Importantly, IFN-γ exposure during activation reduced the cytotoxicity of human-origin type 1 diabetes–relevant autoreactive CD8+ T cells. Collectively, these results indicate that rather than marking the most proinflammatory lymphocytes in diabetes development, IFN-γ production could represent an attempted limitation of pathogenic CD8+ T-cell activation. Thus, great care should be taken when designing possible diabetic intervention approaches modulating IFN-γ production.
- Published
- 2016
- Full Text
- View/download PDF
29. Dysregulation of the PA/PAI System in Pulmonary Disease (ARDS and Fibrosis)
- Author
-
Harold A. Chapman
- Subjects
medicine.medical_specialty ,ARDS ,business.industry ,Fibrosis ,Internal medicine ,Pulmonary disease ,Medicine ,business ,medicine.disease ,Gastroenterology - Published
- 2019
- Full Text
- View/download PDF
30. Collagen promotes anti-PD-1/PD-L1 resistance in cancer through LAIR1-dependent CD8
- Author
-
David H, Peng, Bertha Leticia, Rodriguez, Lixia, Diao, Limo, Chen, Jing, Wang, Lauren A, Byers, Ying, Wei, Harold A, Chapman, Mitsuo, Yamauchi, Carmen, Behrens, Gabriela, Raso, Luisa Maren Solis, Soto, Edwin Roger Parra, Cuentes, Ignacio I, Wistuba, Jonathan M, Kurie, and Don L, Gibbons
- Subjects
Male ,Lung Neoplasms ,Programmed Cell Death 1 Receptor ,Datasets as Topic ,Adenocarcinoma of Lung ,Mice, Transgenic ,CD8-Positive T-Lymphocytes ,B7-H1 Antigen ,Proto-Oncogene Proteins p21(ras) ,Carcinoma, Lewis Lung ,Mice ,Antineoplastic Agents, Immunological ,Cell Line, Tumor ,Biomarkers, Tumor ,Animals ,Humans ,RNA-Seq ,Receptors, Immunologic ,Hepatitis A Virus Cellular Receptor 2 ,Lung ,Protein Tyrosine Phosphatase, Non-Receptor Type 6 ,Extracellular Matrix ,Disease Models, Animal ,HEK293 Cells ,Drug Resistance, Neoplasm ,Gene Knockdown Techniques ,Female ,Amino Acid Oxidoreductases ,Collagen - Abstract
Tumor extracellular matrix has been associated with drug resistance and immune suppression. Here, proteomic and RNA profiling reveal increased collagen levels in lung tumors resistant to PD-1/PD-L1 blockade. Additionally, elevated collagen correlates with decreased total CD8
- Published
- 2019
31. Procoagulant Activity and Lung Disease
- Author
-
J. S. Munger, David A. Waltz, and Harold A. Chapman
- Subjects
Lung disease ,business.industry ,Immunology ,Medicine ,business - Published
- 2019
- Full Text
- View/download PDF
32. Secretion of leukotrienes by senescent lung fibroblasts promotes pulmonary fibrosis
- Author
-
Alexis Valdovinos, Cheresa Calhoun, Judith Campisi, Ying Wei, Christopher D. Wiley, Carlos A. Castellanos, Julia R. Jackson, Sonnet S. Davis, Richard Ruan, Claude Jourdan Le Saux, Fatouma Alimirah, Alexis N. Brumwell, Arvind Ramanathan, and Harold A. Chapman
- Subjects
0301 basic medicine ,Male ,Leukotrienes ,Primary Cell Culture ,Prostaglandin ,Cell Line ,03 medical and health sciences ,Idiopathic pulmonary fibrosis ,chemistry.chemical_compound ,Bleomycin ,Mice ,0302 clinical medicine ,Fibrosis ,Pulmonary fibrosis ,medicine ,Animals ,Humans ,Secretion ,Lipoxygenase Inhibitors ,Senolytic ,Lung ,Cellular Senescence ,Arachidonate 5-Lipoxygenase ,medicine.diagnostic_test ,Gene Expression Profiling ,General Medicine ,respiratory system ,Fibroblasts ,medicine.disease ,Idiopathic Pulmonary Fibrosis ,Disease Models, Animal ,030104 developmental biology ,medicine.anatomical_structure ,Bronchoalveolar lavage ,chemistry ,Gene Expression Regulation ,030220 oncology & carcinogenesis ,Culture Media, Conditioned ,Cancer research ,Disease Progression ,Prostaglandins ,Bronchoalveolar Lavage Fluid ,Signal Transduction ,Research Article - Abstract
Accumulation of senescent cells is associated with the progression of pulmonary fibrosis, but mechanisms accounting for this linkage are not well understood. To explore this issue, we investigated whether a class of biologically active profibrotic lipids, the leukotrienes (LT), is part of the senescence-associated secretory phenotype. The analysis of conditioned medium (CM), lipid extracts, and gene expression of LT biosynthesis enzymes revealed that senescent cells secreted LT, regardless of the origin of the cells or the modality of senescence induction. The synthesis of LT was biphasic and followed by antifibrotic prostaglandin (PG) secretion. The LT-rich CM of senescent lung fibroblasts (IMR-90) induced profibrotic signaling in naive fibroblasts, which were abrogated by inhibitors of ALOX5, the principal enzyme in LT biosynthesis. The bleomycin-induced expression of genes encoding LT and PG synthases, level of cysteinyl LT in the bronchoalveolar lavage, and overall fibrosis were reduced upon senescent cell removal either in a genetic mouse model or after senolytic treatment. Quantification of ALOX5(+) cells in lung explants obtained from idiopathic pulmonary fibrosis (IPF) patients indicated that half of these cells were also senescent (p16(Ink4a+)). Unlike human fibroblasts from unused donor lungs made senescent by irradiation, senescent IPF fibroblasts secreted LTs but failed to synthesize PGs. This study demonstrates for the first time to our knowledge that senescent cells secrete functional LTs, significantly contributing to the LT pool known to cause or exacerbate IPF.
- Published
- 2019
33. Urine Proteomics Identifies Novel Biomarkers of IPF Disease Progression and Resolution
- Author
-
M. Rexhepaj, Harold A. Chapman, S. Turner, L.P. Hariri, N. Dupuis, Ying Wei, Paul J. Wolters, M. Decaris, J.R. Greenland, Joyce S. Lee, and J. Vowinckel
- Subjects
Resolution (mass spectrometry) ,business.industry ,Disease progression ,Medicine ,Urine ,Computational biology ,Proteomics ,business - Published
- 2019
- Full Text
- View/download PDF
34. Contextual cues from cancer cells govern cancer-associated fibroblast heterogeneity
- Author
-
Barbara Mino, Young Ho Ahn, Neus Bota-Rabassedas, Edwin R. Parra, Chenghang Zong, Daniel W. Sazer, Jonathan M. Kurie, Joshua J.A. Firestone, Wenjian Cao, B. Leticia Rodriguez, Sieun Lee, Jing Wang, Priyam Banerjee, Jiayi Luo, Xiaochao Tan, Xin Liu, Luisa M. Solis, Jordan S. Miller, Jacob Albritton, Maria Gabriela Raso, Gregory D. Longmore, Pamela Villalobos, Chad J. Creighton, Hou Fu Guo, Ignacio I. Wistuba, Yichi Niu, Jiang Yu, William K. Russell, Don L. Gibbons, Yuanxin Xi, Michael Weiger, Harold A. Chapman, Jaime Rodriguez-Canales, and Kuanwei Sheng
- Subjects
0301 basic medicine ,Male ,Lung Neoplasms ,cancer-associated fibroblast ,Cell Communication ,Metastasis ,Mice ,0302 clinical medicine ,Cancer-Associated Fibroblasts ,Cell Movement ,Tumor Microenvironment ,Biology (General) ,microRNA ,EMT ,Kidney Neoplasms ,Gene Expression Regulation, Neoplastic ,medicine.anatomical_structure ,Adenocarcinoma ,Signal Transduction ,Stromal cell ,Epithelial-Mesenchymal Transition ,QH301-705.5 ,Adenocarcinoma of Lung ,Mice, Transgenic ,Biology ,General Biochemistry, Genetics and Molecular Biology ,Article ,03 medical and health sciences ,Discoidin Domain Receptor 2 ,Cell Line, Tumor ,Alpha-Globulins ,medicine ,metastasis ,Animals ,Humans ,Fibroblast ,Transcription factor ,Cell Proliferation ,Tumor microenvironment ,Gene Expression Profiling ,Zinc Finger E-box-Binding Homeobox 1 ,Epithelial Cells ,Mesenchymal Stem Cells ,medicine.disease ,lung cancer ,030104 developmental biology ,Cancer cell ,Cancer research ,030217 neurology & neurosurgery - Abstract
SUMMARY Cancer cells function as primary architects of the tumor microenvironment. However, the molecular features of cancer cells that govern stromal cell phenotypes remain unclear. Here, we show that cancer-associated fibroblast (CAF) heterogeneity is driven by lung adenocarcinoma (LUAD) cells at either end of the epithelial-to-mesenchymal transition (EMT) spectrum. LUAD cells that have high expression of the EMT-activating transcription factor ZEB1 reprogram CAFs through a ZEB1-dependent secretory program and direct CAFs to the tips of invasive projections through a ZEB1-driven CAF repulsion process. The EMT, in turn, sensitizes LUAD cells to pro-metastatic signals from CAFs. Thus, CAFs respond to contextual cues from LUAD cells to promote metastasis., In brief Bota-Rabassedas et al. show that EMT in lung adenocarcinoma cells activates a secretory process that governs CAF heterogeneity and, in turn, sensitizes lung adenocarcinoma cells to pro-metastatic signals from CAFs. Thus, EMT positions lung adenocarcinoma cells at the apex of a signaling hierarchy in the tumor microenvironment., Graphical Abstract
- Published
- 2021
35. Diabetes
- Author
-
Estela Rosell-Mases, Jeremy J. Racine, Jorge Carrascal, Berta Arpa, Caroline M. Leeth, Qiming Wang, David V. Serreze, Leire Egia-Mendikute, Thomas Stratmann, Jorge Carrillo, Joan Verdaguer, Jeremy J. Ratiu, Harold D. Chapman, and Animal and Poultry Sciences
- Subjects
0301 basic medicine ,INSULITIS ,SUBSETS ,Endocrinology, Diabetes and Metabolism ,Nod ,Biology ,Endocrinology & Metabolism ,ANTIGEN-PRESENTING CELLS ,INITIATION ,03 medical and health sciences ,0302 clinical medicine ,Antigen ,Internal Medicine ,medicine ,TOLERANCE ,Antigen-presenting cell ,DESTRUCTION ,NOD mice ,INTERMEDIATE-FILAMENT PROTEIN ,Pancreatic islets ,ANTIBODY PREVENTS ,Peripherin ,medicine.disease ,Tolerance induction ,030104 developmental biology ,medicine.anatomical_structure ,Immunology ,Immunology and Transplantation ,DEPLETION ,ANERGY ,Insulitis ,030215 immunology - Abstract
While the autoimmune destruction of pancreatic ß-cells underlying type 1 diabetes (1D) development is ultimately mediated by T-cells in NOD mice and also likely humans, B-lymphocytes play an additional key pathogenic role. It appears expression of plasma membrane bound immunoglobulin (Ig) molecules that efficiently capture ß-cell antigens allows autoreactive B-lymphocytes bypassing normal tolerance induction processes to be the subset of antigen presenting cells most efficiently activating diabetogenic T-cells. NOD mice transgenically expressing Ig molecules recognizing antigens that are (insulin) or not (hen egg lysozyme; HEL) expressed by ß-cells have proven useful in dissecting the developmental basis of diabetogenic B-lymphocytes. However, these transgenic Ig specificities were originally selected for their ability to recognize insulin or HEL as foreign, rather than autoantigens. Thus, we generated and characterized NOD mice transgenically expressing an Ig molecule representative of a large proportion of naturally occurring islet-infiltrating B-lymphocytes in NOD mice recognizing the neuronal antigen peripherin. Transgenic peripherin autoreactive B-lymphocytes infiltrate NOD pancreatic islets, acquire an activated proliferative phenotype, and potently support accelerated T1D development. These results support the concept of neuronal autoimmunity as a pathogenic feature of T1D, and targeting such responses could ultimately provide an effective disease intervention approach.
- Published
- 2016
- Full Text
- View/download PDF
36. A Hypermorphic Allele Contributes to Impaired Thymic Deletion of Autoreactive Diabetogenic CD8 T Cells in NOD Mice
- Author
-
Jennifer Allocco, Tim Stearns, Aron M. Geurts, Yi-Guang Chen, Maximiliano Presa, Ingo Schmitz, Deanna J. Lamont, Harold D. Chapman, David V. Serreze, Vishal Kumar Sarsani, Jennifer R. Dwyer, Jeremy J. Ratiu, Jeremy J. Racine, and HZI,Helmholtz-Zentrum für Infektionsforschung GmbH, Inhoffenstr. 7,38124 Braunschweig, Germany.
- Subjects
0301 basic medicine ,biology ,T cell ,Immunology ,Major histocompatibility complex ,Molecular biology ,NFKBID ,Clonal deletion ,03 medical and health sciences ,030104 developmental biology ,0302 clinical medicine ,medicine.anatomical_structure ,MHC class I ,biology.protein ,medicine ,Immunology and Allergy ,Cytotoxic T cell ,CD8 ,030215 immunology ,NOD mice - Abstract
In both NOD mice and humans, the development of type 1 diabetes (T1D) is dependent in part on autoreactive CD8+ T cells recognizing pancreatic β cell peptides presented by often quite common MHC class I variants. Studies in NOD mice previously revealed that the common H2-Kd and/or H2-Db class I molecules expressed by this strain aberrantly lose the ability to mediate the thymic deletion of pathogenic CD8+ T cell responses through interactions with T1D susceptibility genes outside the MHC. A gene(s) mapping to proximal chromosome 7 was previously shown to be an important contributor to the failure of the common class I molecules expressed by NOD mice to mediate the normal thymic negative selection of diabetogenic CD8+ T cells. Using an inducible model of thymic negative selection and mRNA transcript analyses, we initially identified an elevated Nfkbid expression variant as a likely NOD-proximal chromosome 7 region gene contributing to impaired thymic deletion of diabetogenic CD8+ T cells. CRISPR/Cas9–mediated genetic attenuation of Nfkbid expression in NOD mice resulted in improved negative selection of autoreactive diabetogenic AI4 and NY8.3 CD8+ T cells. These results indicated that allelic variants of Nfkbid contribute to the efficiency of intrathymic deletion of diabetogenic CD8+ T cells. However, although enhancing thymic deletion of pathogenic CD8+ T cells, ablating Nfkbid expression surprisingly accelerated T1D onset that was associated with numeric decreases in both regulatory T and B lymphocytes in NOD mice.
- Published
- 2018
37. Small molecule inhibition of IRE1α kinase/RNase has anti-fibrotic effects in the lung
- Author
-
Alexis N. Brumwell, Dustin J. Maly, Harold A. Chapman, Bradley J. Backes, Vincent C. Auyeung, Dean Sheppard, Rajarshi Ghosh, Feroz R. Papa, Maike Thamsen, and Gayani Perara
- Subjects
0301 basic medicine ,General Science & Technology ,Science ,Apoptosis ,Protein Serine-Threonine Kinases ,Bleomycin ,Cell Line ,03 medical and health sciences ,Mice ,Idiopathic pulmonary fibrosis ,chemistry.chemical_compound ,0302 clinical medicine ,Fibrosis ,Endoribonucleases ,Pulmonary fibrosis ,medicine ,Animals ,Lung ,Protein Kinase Inhibitors ,Multidisciplinary ,Kinase ,Endoplasmic reticulum ,Protein-Serine-Threonine Kinases ,Endoplasmic Reticulum Stress ,medicine.disease ,030104 developmental biology ,medicine.anatomical_structure ,chemistry ,030220 oncology & carcinogenesis ,Alveolar Epithelial Cells ,Unfolded Protein Response ,Cancer research ,Unfolded protein response ,Medicine ,Signal transduction - Abstract
Endoplasmic reticulum stress (ER stress) has been implicated in the pathogenesis of idiopathic pulmonary fibrosis (IPF), a disease of progressive fibrosis and respiratory failure. ER stress activates a signaling pathway called the unfolded protein response (UPR) that either restores homeostasis or promotes apoptosis. The bifunctional kinase/RNase IRE1α is a UPR sensor that promotes apoptosis if ER stress remains high (i.e., a “terminal” UPR). Using multiple small molecule inhibitors against IRE1α, we show that ER stress-induced apoptosis of murine alveolar epithelial cells can be mitigated in vitro. In vivo, we show that bleomycin exposure to murine lungs causes early ER stress to activate IRE1α and the terminal UPR prior to development of pulmonary fibrosis. Small-molecule IRE1α kinase-inhibiting RNase attenuators (KIRAs) that we developed were used to evaluate the importance of IRE1α activation in bleomycin-induced pulmonary fibrosis. One such KIRA—KIRA7—provided systemically to mice at the time of bleomycin exposure decreases terminal UPR signaling and prevents lung fibrosis. Administration of KIRA7 14 days after bleomycin exposure even promoted the reversal of established fibrosis. Finally, we show that KIRA8, a nanomolar-potent, monoselective KIRA compound derived from a completely different scaffold than KIRA7, likewise promoted reversal of established fibrosis. These results demonstrate that IRE1α may be a promising target in pulmonary fibrosis and that kinase inhibitors of IRE1α may eventually be developed into efficacious anti-fibrotic drugs.
- Published
- 2018
- Full Text
- View/download PDF
38. Photosynthesis in the potato, Solanum tuberosum L
- Author
-
Harold W. Chapman
- Subjects
Botany ,Biology ,Photosynthesis ,Solanum tuberosum - Published
- 2018
- Full Text
- View/download PDF
39. Idiopathic Pulmonary Fibrosis: Cell Death and Inflammation Revisited
- Author
-
Claude Jourdan Le Saux and Harold A. Chapman
- Subjects
0301 basic medicine ,Pulmonary and Respiratory Medicine ,Inflammation ,Programmed cell death ,Pathology ,medicine.medical_specialty ,Cell Death ,business.industry ,Clinical Biochemistry ,Apoptosis ,Cell Biology ,medicine.disease ,Idiopathic Pulmonary Fibrosis ,03 medical and health sciences ,Idiopathic pulmonary fibrosis ,030104 developmental biology ,Text mining ,Alveolar Epithelial Cells ,medicine ,Humans ,medicine.symptom ,business ,Molecular Biology - Published
- 2018
40. TGF-β1 Signaling and Tissue Fibrosis
- Author
-
Dean Sheppard, Harold A. Chapman, and Kevin K. Kim
- Subjects
0301 basic medicine ,Integrins ,Epithelial-Mesenchymal Transition ,Physiological ,Apoptosis ,Biology ,Models, Biological ,General Biochemistry, Genetics and Molecular Biology ,Feedback ,Extracellular matrix ,Transforming Growth Factor beta1 ,03 medical and health sciences ,Mice ,0302 clinical medicine ,Fibrosis ,Models ,medicine ,Animals ,Humans ,Epithelial–mesenchymal transition ,Pathological ,Cell Proliferation ,Feedback, Physiological ,Wound Healing ,Anatomy ,medicine.disease ,Biological ,Cell biology ,030104 developmental biology ,PERSPECTIVES ,Tgf β1 signaling ,030220 oncology & carcinogenesis ,Tissue fibrosis ,Collagen ,Signal transduction ,Wound healing ,Signal Transduction - Abstract
© 2018 Cold Spring Harbor Laboratory Press; all rights reserved. Activation of TGF-β1 initiates a program of temporary collagen accumulation important to wound repair in many organs. However, the outcome of temporary extracellular matrix strengthening all too frequently morphs into progressive fibrosis, contributing to morbidity and mortality worldwide. To avoid this maladaptive outcome, TGF-β1 signaling is regulated at numerous levels and intimately connected to feedback signals that limit accumulation. Here, we examine the current understanding of the core functions of TGF-β1 in promoting collagen accumulation, parallel pathways that promote physiological repair, and pathological triggers that tip the balance toward progressive fibrosis. Implicit in better understanding of these processes is the identification of therapeutic opportunities that will need to be further advanced to limit or reverse organ fibrosis.
- Published
- 2018
- Full Text
- View/download PDF
41. A Hypermorphic
- Author
-
Maximiliano, Presa, Jeremy J, Racine, Jennifer R, Dwyer, Deanna J, Lamont, Jeremy J, Ratiu, Vishal Kumar, Sarsani, Yi-Guang, Chen, Aron, Geurts, Ingo, Schmitz, Timothy, Stearns, Jennifer, Allocco, Harold D, Chapman, and David V, Serreze
- Subjects
Polymorphism, Genetic ,Clonal Deletion ,Cell Differentiation ,Thymus Gland ,CD8-Positive T-Lymphocytes ,Autoantigens ,Article ,Disease Models, Animal ,Mice ,Diabetes Mellitus, Type 1 ,Mice, Inbred NOD ,Animals ,Humans ,I-kappa B Proteins ,Disease Susceptibility ,Alleles ,Cells, Cultured ,Chromosomes, Human, Pair 7 - Abstract
In both NOD mice and humans, the development of type 1 diabetes (T1D) is dependent in part on autoreactive CD8(+) T-cells recognizing pancreatic ß-cell peptides presented by often quite common MHC class I variants. Studies in NOD mice previously revealed the common H2-K(d) and/or H2-D(b) class I molecules expressed by this strain aberrantly lose the ability to mediate the thymic deletion of pathogenic CD8(+) T-cell responses through interactions with T1D susceptibility (Idd) genes outside the MHC. A gene(s) mapping to proximal Chromosome 7 was previously shown to be an important contributor to the failure of the common class I molecules expressed by NOD mice to mediate the normal thymic negative selection of diabetogenic CD8(+) T-cells. Using an inducible model of thymic negative selection and mRNA transcript analyses we initially identified an elevated Nfkbid expression variant as a likely NOD proximal Chromosome 7 region gene contributing to impaired thymic deletion of diabetogenic CD8(+) T-cells. CRISPR/Cas9-mediated genetic attenuation of Nfkbid expression in NOD mice resulted in improved negative selection of autoreactive diabetogenic AI4 and NY8.3 CD8(+) T-cells. These results indicated allelic variants of Nfkbid contribute to the efficiency of intrathymic deletion of diabetogenic CD8(+) T-cells. However, while enhancing thymic deletion of pathogenic CD8(+) T-cells, ablating Nfkbid expression surprisingly accelerated T1D onset in NOD mice that was associated with numeric decreases in both regulatory T- and B-lymphocytes (Tregs/Bregs).
- Published
- 2018
42. HLA-B*39:06 Efficiently Mediates Type 1 Diabetes in a Mouse Model Incorporating Reduced Thymic Insulin Expression1
- Author
-
Harold D. Chapman, Jeremy J. Racine, Riyasat Ali, Jennifer Schloss, Teresa P. DiLorenzo, and David V. Serreze
- Subjects
0303 health sciences ,Type 1 diabetes ,Insulin ,medicine.medical_treatment ,Pancreatic islets ,T cell ,Human leukocyte antigen ,Biology ,medicine.disease ,Major histocompatibility complex ,03 medical and health sciences ,0302 clinical medicine ,medicine.anatomical_structure ,Immunology ,medicine ,biology.protein ,Cytotoxic T cell ,030304 developmental biology ,030215 immunology ,NOD mice - Abstract
Type 1 diabetes (T1D) is characterized by T cell-mediated destruction of the insulin-producing βcells of the pancreatic islets. Among the loci associated with T1D risk, those most predisposing are found in the MHC region. HLA-B*39:06 is the most predisposing class I MHC allele and is associated with an early age of onset. To establish an NOD mouse model for the study of HLA-B*39:06, we expressed it in the absence of murine class I MHC. HLA-B*39:06 was able to mediate the development of CD8 T cells, support lymphocytic infiltration of the islets, and confer T1D susceptibility. Because reduced thymic insulin expression is associated with increased T1D risk in patients, we incorporated this in our model as well, finding that HLA-B*39:06-transgenic NOD mice with reduced thymic insulin expression have an earlier age of disease onset and a higher overall prevalence as compared to littermates with typical thymic insulin expression. This was despite virtually indistinguishable blood insulin levels, T cell subset percentages, and TCR Vβ family usage, indicating that reduced thymic insulin expression does not impact T cell development on a global scale. Rather, we propose that it allows the thymic escape of insulin-reactive HLA-B*39:06-restricted T cells which participate in β cell destruction. We also found that in mice expressing either HLA-B*39:06 or HLA-A*02:01 in the absence of murine class I MHC, HLA transgene identity alters TCR Vβ usage, which may contribute to varying diabetogenic CD8 T cell repertoires in the presence of different HLA class I alleles.
- Published
- 2018
- Full Text
- View/download PDF
43. A hypermorphic Nfkbid allele represents an Idd7 locus gene contributing to impaired thymic deletion of autoreactive diabetogenic CD8+ T-cells in NOD mice
- Author
-
Jennifer R. Dwyer, Jeremy J. Racine, Tim Stearns, Jeremy J. Ratiu, Maximiliano Presa, Vishal Kumar Sarsani, Jennifer Allocco, David V. Serreze, Ingo Schmitz, Deanna J. Lamont, Aron M. Geurts, Yi-Guang Chen, and Harold D. Chapman
- Subjects
0303 health sciences ,biology ,Nod ,Major histocompatibility complex ,03 medical and health sciences ,Negative selection ,0302 clinical medicine ,MHC class I ,Immunology ,biology.protein ,Cytotoxic T cell ,Gene ,CD8 ,030304 developmental biology ,030215 immunology ,NOD mice - Abstract
In both NOD mice and humans, the development of type 1 diabetes (T1D) is dependent in part on autoreactive CD8+ T-cells recognizing pancreatic ß-cell peptides presented by often quite common MHC class I variants. Studies in NOD mice previously revealed the common H2-Kd and/or H2-Db class I molecules expressed by this strain acquire an aberrant ability to mediate pathogenic CD8+ T-cell responses through interactions with T1D susceptibility (Idd) genes outside the MHC. A gene(s) mapping to the Idd7 locus on proximal Chromosome 7 was previously shown to be an important contributor to the failure of the common class I molecules expressed by NOD mice to mediate the normal thymic negative selection of diabetogenic CD8+ T-cells. Using an inducible model of thymic negative selection and mRNA transcript analyses we initially identified an elevated Nfkbid expression variant is likely an NOD Idd7 region gene contributing to impaired thymic deletion of diabetogenic CD8+ T-cells. CRISPR/Cas9-mediated genetic attenuation of Nfkbid expression in NOD mice resulted in improved negative selection of autoreactive diabetogenic AI4 and NY8.3 CD8+ T-cells. These results indicated allelic variants of Nfkbid represent an Idd7 gene contributing to the efficiency of intrathymic deletion of diabetogenic CD8+ T-cells. However, while enhancing thymic deletion of pathogenic CD8+ T-cells, ablation of Nfkbid expression surprisingly accelerated T1D onset in NOD mice likely at least in part by numerically decreasing regulatory T- and B-lymphocytes (Tregs/Bregs), thereby reducing their peripheral immunosuppressive effects.
- Published
- 2018
- Full Text
- View/download PDF
44. Lineage-negative progenitors mobilize to regenerate lung epithelium after major injury
- Author
-
Jeffrey E. Gotts, Mark R. Looney, Fengchun Liu, Kevin Tan, Andrew E. Vaughan, Ying Xi, Victor M. Tan, Michael A. Matthay, Barbara Treutlein, Alexis N. Brumwell, Jason R. Rock, Douglas Brownfield, and Harold A. Chapman
- Subjects
Male ,Cellular differentiation ,Notch signaling pathway ,Cell Separation ,Biology ,Lung injury ,Article ,Bleomycin ,Mice ,03 medical and health sciences ,0302 clinical medicine ,Orthomyxoviridae Infections ,Re-Epithelialization ,Animals ,Humans ,Regeneration ,Cell Lineage ,Progenitor cell ,Lung ,Cell Proliferation ,030304 developmental biology ,0303 health sciences ,Multidisciplinary ,Receptors, Notch ,Cysts ,Stem Cells ,Regeneration (biology) ,Lineage markers ,Epithelial Cells ,Lung Injury ,Phosphoproteins ,Cell biology ,030220 oncology & carcinogenesis ,Immunology ,Trans-Activators ,Keratin-5 ,Female ,Stem cell ,Signal Transduction ,Stem Cell Transplantation ,Adult stem cell - Abstract
Broadly, tissue regeneration is achieved in two ways: by proliferation of common differentiated cells and/or by deployment of specialized stem/progenitor cells. Which of these pathways applies is both organ- and injury-specific. Current models in the lung posit that epithelial repair can be attributed to cells expressing mature lineage markers. By contrast, here we define the regenerative role of previously uncharacterized, rare lineage-negative epithelial stem/progenitor (LNEP) cells present within normal distal lung. Quiescent LNEPs activate a ΔNp63 (a p63 splice variant) and cytokeratin 5 remodelling program after influenza or bleomycin injury in mice. Activated cells proliferate and migrate widely to occupy heavily injured areas depleted of mature lineages, at which point they differentiate towards mature epithelium. Lineage tracing revealed scant contribution of pre-existing mature epithelial cells in such repair, whereas orthotopic transplantation of LNEPs, isolated by a definitive surface profile identified through single-cell sequencing, directly demonstrated the proliferative capacity and multipotency of this population. LNEPs require Notch signalling to activate the ΔNp63 and cytokeratin 5 program, and subsequent Notch blockade promotes an alveolar cell fate. Persistent Notch signalling after injury led to parenchymal 'micro-honeycombing' (alveolar cysts), indicative of failed regeneration. Lungs from patients with fibrosis show analogous honeycomb cysts with evidence of hyperactive Notch signalling. Our findings indicate that distinct stem/progenitor cell pools repopulate injured tissue depending on the extent of the injury, and the outcomes of regeneration or fibrosis may depend in part on the dynamics of LNEP Notch signalling.
- Published
- 2014
- Full Text
- View/download PDF
45. Innate Antiviral Host Defense Attenuates TGF-β Function through IRF3-Mediated Suppression of Smad Signaling
- Author
-
Ying Xi, Dan Du, Daqi Xu, Pinglong Xu, Samantha L. Bailey-Bucktrout, Jeffrey A. Bluestone, Weiwen Xiang, Qian Zhang, Andrew C. Melton, Dean Sheppard, Rik Derynck, Jianming Liu, and Harold A. Chapman
- Subjects
Transcriptional Activation ,Epithelial-Mesenchymal Transition ,Transcription, Genetic ,1.1 Normal biological development and functioning ,T-Lymphocytes ,Cellular differentiation ,SMAD ,Biology ,Inbred C57BL ,Medical and Health Sciences ,Sendai virus ,T-Lymphocytes, Regulatory ,Immune tolerance ,Vaccine Related ,Mice ,Genetic ,Underpinning research ,Transforming Growth Factor beta ,2.1 Biological and endogenous factors ,Innate ,Animals ,Humans ,Gene silencing ,Smad3 Protein ,Aetiology ,Molecular Biology ,Cancer ,Regulation of gene expression ,Effector ,Inflammatory and immune system ,Immunity ,Cell Differentiation ,Hep G2 Cells ,Cell Biology ,Biological Sciences ,biochemical phenomena, metabolism, and nutrition ,Regulatory ,Immunity, Innate ,Cell biology ,Mice, Inbred C57BL ,Infectious Diseases ,Emerging Infectious Diseases ,HEK293 Cells ,Interferon Regulatory Factor-3 ,Signal transduction ,IRF3 ,Transcription ,Developmental Biology ,Signal Transduction - Abstract
© 2014 Elsevier Inc. TGF-β signaling is essential in many processes, including immune surveillance, and its dysregulation controls various diseases, including cancer, fibrosis, and inflammation. Studying the innate host defense, which functions in most cell types, we found that RLR signaling represses TGF-β responses. This regulation is mediated by activated IRF3, using a dual mechanism of IRF3-directed suppression. Activated IRF3 interacts with Smad3, thus inhibiting TGF-β-induced Smad3 activation and, in the nucleus, disrupts functional Smad3 transcription complexes by competing with coregulators. Consequently, IRF3 activation by innate antiviral signaling represses TGF-β-induced growth inhibition, gene regulation and epithelial-mesenchymal transition, and the generation of Treg effector lymphocytes from naive CD4+lymphocytes. Conversely, silencing IRF3 expression enhances epithelial-mesenchymal transition, TGF-β-induced Treg cell differentiation upon virus infection, and Treg cell generation invivo. Wepresent a mechanism of regulation of TGF-β signaling by the antiviral defense, with evidence for its role in immune tolerance and cancer cell behavior.
- Published
- 2014
- Full Text
- View/download PDF
46. Persistent Pathology in Influenza-Infected Mouse Lungs
- Author
-
Cindy M Kanegai, Jeffrey E. Gotts, Kirk D. Jones, Ying Xi, Gorica Amidzic, Jason R. Rock, Harold A. Chapman, Andrew J. Lechner, Ian Driver, Andrew E. Vaughan, and Matthew Donne
- Subjects
0301 basic medicine ,Pulmonary and Respiratory Medicine ,Extramural ,business.industry ,Clinical Biochemistry ,MEDLINE ,Respiratory Mucosa ,Cell Biology ,medicine.disease_cause ,Virology ,Pulmonary Alveoli ,Mice ,03 medical and health sciences ,Influenza A Virus, H1N1 Subtype ,030104 developmental biology ,0302 clinical medicine ,Text mining ,Orthomyxoviridae Infections ,030228 respiratory system ,Correspondence ,Influenza A virus ,medicine ,Animals ,business ,Molecular Biology - Published
- 2016
- Full Text
- View/download PDF
47. Antibiotic-associated Manipulation of the Gut Microbiota and Phenotypic Restoration in NOD Mice
- Author
-
James R, Fahey, Bonnie L, Lyons, Haiyan L, Olekszak, Anthony J, Mourino, Jeremy J, Ratiu, Jeremy J, Racine, Harold D, Chapman, David V, Serreze, Dina L, Baker, and N Ken, Hendrix
- Subjects
Time Factors ,Mouse Models ,Th1 Cells ,Gram-Positive Bacteria ,Anti-Bacterial Agents ,Gastrointestinal Microbiome ,Intestines ,Disease Models, Animal ,Feces ,Diabetes Mellitus, Type 1 ,Phenotype ,Mice, Inbred NOD ,Host-Pathogen Interactions ,Animals ,Th17 Cells ,Ampicillin ,Genetic Predisposition to Disease ,Animal Husbandry ,Decontamination ,Gram-Positive Bacterial Infections ,Environmental Monitoring - Abstract
Segmented filamentous bacterium (SFB) a gram-positive, anaerobic, and intestinal commensal organism directly influences the development of Th17 helper cells in the small intestine of mice. In NOD mice, SFB colonization interferes with the development of type 1 diabetes (T1D), a T-cell–mediated autoimmune disease, suggesting that SFB may influence Th17 cells to inhibit Th1 populations associated with the anti-β-cell immune response. This effect is a serious concern for investigators who use NOD mice for diabetes research because the expected incidence of disease decreases markedly when they are colonized by SFB. A room housing mice for T1D studies at The Jackson Laboratory was determined by fecal PCR testing to have widespread SFB colonization of multiple NOD strains after a steady decline in the incidence of T1D was noted. Rederivation of all NOD-related mouse strains was not feasible; therefore an alternative treatment using antibiotics to eliminate SFB from colonized mice was undertaken. After antibiotic treatment, soiled bedding from NOD mouse strains housed in SFB-free high-health–status production barrier rooms was used to reintroduce the gastrointestinal microbiota. Over the past 16 mo since treating the mice and disinfecting the mouse room, regular PCR testing has shown that no additional SFB colonization of mice has occurred, and the expected incidence of T1D has been reestablished in the offspring of treated mice.
- Published
- 2017
48. Hypoxia-Inducible Factor 1α Signaling Promotes Repair of the Alveolar Epithelium after Acute Lung Injury
- Author
-
Rubin M. Tuder, Yoko Ito, Nicole L. Jansing, Robert J. Mason, Rachel L. Zemans, Jazalle McClendon, Sean P. Colgan, Aneta Gandjeva, David W. H. Riches, Harold A. Chapman, Elizabeth F. Redente, and Aftab Ahmad
- Subjects
0301 basic medicine ,Vascular Endothelial Growth Factor A ,Pathology ,Regenerative Medicine ,Medical and Health Sciences ,chemistry.chemical_compound ,Mice ,Receptors ,2.1 Biological and endogenous factors ,Aetiology ,Lung ,Acute Respiratory Distress Syndrome ,Regular Article ,respiratory system ,Cell biology ,Vascular endothelial growth factor ,medicine.anatomical_structure ,Hypoxia-inducible factors ,Respiratory ,Hypoxia-Inducible Factor 1 ,Signal Transduction ,Receptors, CXCR4 ,medicine.medical_specialty ,Stromal cell ,Alveolar Epithelium ,Acute Lung Injury ,Biology ,Lung injury ,alpha Subunit ,Permeability ,Pathology and Forensic Medicine ,Cell Line ,03 medical and health sciences ,Rare Diseases ,medicine ,Animals ,Cell Proliferation ,Basement membrane ,CXCR4 ,Wound Healing ,Cell growth ,Animal ,Hypoxia-Inducible Factor 1, alpha Subunit ,Chemokine CXCL12 ,Rats ,Pulmonary Alveoli ,Disease Models, Animal ,030104 developmental biology ,chemistry ,Cell culture ,Alveolar Epithelial Cells ,Disease Models - Abstract
During the acute respiratory distress syndrome, epithelial cells, primarily alveolar type (AT) I cells, die and slough off, resulting in enhanced permeability. ATII cells proliferate and spread onto the denuded basement membrane to reseal the barrier. Repair of the alveolar epithelium is critical for clinical recovery; however, mechanisms underlying ATII cell proliferation and spreading are not well understood. We hypothesized that hypoxia-inducible factor (HIF)1α promotes proliferation and spreading of ATII cells during repair after lung injury. Mice were treated with lipopolysaccharide or hydrochloric acid. HIF activation in ATII cells after injury was demonstrated by increased luciferase activity in oxygen degradation domain-Luc (HIF reporter) mice and expression of the HIF1α target gene GLUT1. ATII cell proliferation during repair was attenuated in ATII cell-specific HIF1α knockout (SftpcCreERT2+/-;HIF1αf/f) mice. The HIF target vascular endothelial growth factor promoted ATII cell proliferation invitro and after lung injury invivo. In the scratch wound assay of cell spreading, HIF stabilization accelerated, whereas HIF1α shRNA delayed wound closure. SDF1 and its receptor, CXCR4, were found to be HIF1α-regulated genes in ATII cells and were up-regulated during lung injury. Stromal cell-derived factor 1/CXCR4 inhibition impaired cell spreading and delayed the resolution of permeability after lung injury. We conclude that HIF1α is activated in ATII cells after lung injury and promotes proliferation and spreading during repair.
- Published
- 2017
49. Urokinase-type Plasminogen Activator Receptor (uPAR) Ligation Induces a Raft-localized Integrin Signaling Switch That Mediates the Hypermotile Phenotype of Fibrotic Fibroblasts
- Author
-
Efrat Harel, Charles S. Craik, Sailaja Paruchuri, Harold A. Chapman, Brian D. Southern, Candece L. Gladson, Ying Wei, Kimberly E. White, Shaik O. Rahaman, Qiang Ding, Tong H. Jin, Mitchell A. Olman, and Lisa M. Grove
- Subjects
Glycobiology and Extracellular Matrices ,Integrin ,Proto-Oncogene Proteins c-fyn ,Severity of Illness Index ,Medical and Health Sciences ,Biochemistry ,Mice ,Cell Movement ,Receptors ,2.1 Biological and endogenous factors ,Src family kinase ,Aetiology ,skin and connective tissue diseases ,Lung ,Lipid raft ,Cells, Cultured ,Microscopy ,Cultured ,biology ,Blotting ,respiratory system ,Biological Sciences ,Urokinase Plasminogen Activator ,Urokinase Receptor ,Respiratory ,Fibroblast ,RNA Interference ,lipids (amino acids, peptides, and proteins) ,biological phenomena, cell phenomena, and immunity ,Signal transduction ,Western ,Integrin alpha5beta1 ,Signal Transduction ,Protein Binding ,Biochemistry & Molecular Biology ,Cells ,Blotting, Western ,Caveolins ,Autoimmune Disease ,Fluorescence ,Receptors, Urokinase Plasminogen Activator ,Focal adhesion ,Membrane Microdomains ,Rare Diseases ,FYN ,Animals ,Humans ,neoplasms ,Molecular Biology ,Lipid Raft ,Cell Biology ,Fibroblasts ,Urokinase-Type Plasminogen Activator ,Fibrosis ,biological factors ,Idiopathic Pulmonary Fibrosis ,respiratory tract diseases ,Fibronectins ,Fibronectin ,Urokinase receptor ,enzymes and coenzymes (carbohydrates) ,Microscopy, Fluorescence ,Shc Signaling Adaptor Proteins ,Chemical Sciences ,biology.protein ,Cancer research - Abstract
The urokinase-type plasminogen activator receptor (uPAR) is a glycosylphosphatidylinositol-linked membrane protein with no cytosolic domain that localizes to lipid raft microdomains. Our laboratory and others have documented that lung fibroblasts from patients with idiopathic pulmonary fibrosis (IPF) exhibit a hypermotile phenotype. This study was undertaken to elucidate the molecular mechanism whereby uPAR ligation with its cognate ligand, urokinase, induces a motile phenotype in human lung fibroblasts. We found that uPAR ligation with the urokinase receptor binding domain (amino-terminal fragment) leads to enhanced migration of fibroblasts on fibronectin in a protease-independent, lipid raft-dependent manner. Ligation of uPAR with the amino-terminal fragment recruited α5β1 integrin and the acylated form of the Src family kinase, Fyn, to lipid rafts. The biological consequences of this translocation were an increase in fibroblast motility and a switch of the integrin-initiated signal pathway for migration away from the lipid raft-independent focal adhesion kinase pathway and toward a lipid raft-dependent caveolin-Fyn-Shc pathway. Furthermore, an integrin homologous peptide as well as an antibody that competes with β1 for uPAR binding have the ability to block this effect. In addition, its relative insensitivity to cholesterol depletion suggests that the interactions of α5β1 integrin and uPAR drive the translocation of α5β1 integrin-acylated Fyn signaling complexes into lipid rafts upon uPAR ligation through protein-protein interactions. This signal switch is a novel pathway leading to the hypermotile phenotype of IPF patient-derived fibroblasts, seen with uPAR ligation. This uPAR dependent, fibrotic matrix-selective, and profibrotic fibroblast phenotype may be amenable to targeted therapeutics designed to ameliorate IPF.
- Published
- 2014
- Full Text
- View/download PDF
50. Nfkbid overexpression in NOD mice elicits complete type 1 diabetes resistance
- Author
-
Jennifer R Dwyer, Jeremy J Racine, Anna Quinlan, Maximiliano Presa, Harold D Chapman, and David V Serreze
- Subjects
Immunology ,Immunology and Allergy - Abstract
Type 1 diabetes (T1D) in both humans and the NOD mouse model is caused by the autoimmune mediated killing of pancreatic β-cells. Increased frequency or activity of autoreactive T cells and failures of regulatory T cells (Tregs) to control autoreactive T cells have both been implicated in T1D etiology. Due to the expression of MHC-I molecules on β-cells, autoreactive CD8+ T cells are likely the ultimate mediators of this destruction. Developing CD8+ T cells undergo selection processes in the thymus whereby most autoreactive clonotypes are eliminated during their development. Fine mapping of a genetic locus associated with defective thymic deletion of autoreactive CD8+ AI4 TCR transgenic (AI4) T cells in NOD mice led to the identification of Nfkbid, a NFκB signal modulator, as contributing to this phenotype. Ablation of Nfkbid in NOD mice by CRISPR/Cas9 results in a decreased frequency of autoreactive CD8+ AI4 T cells and an unexpected acceleration of T1D incidence, associated with reductions in the frequency and functional capacity of peripheral Tregs. Transgenic overexpression of Nfkbid in NOD mice paradoxically results in further decreased levels of circulating autoreactive CD8+ AI4 T cells. However, transgenic elevation of Nfkbid expression also increased the frequency and functional capacity of peripheral Tregs with a striking complete absence of T1D development in these mice. Studies are ongoing to define the mechanisms of T1D resistance mediated by transgenic overexpression of the transcriptional co-factor Nfkbid.
- Published
- 2019
- Full Text
- View/download PDF
Catalog
Discovery Service for Jio Institute Digital Library
For full access to our library's resources, please sign in.