1
|
Jackson JW, Frederick C Streich, Pal A, Coricor G, Boston C, Brueckner CT, Canonico K, Chapron C, Cote S, Dagbay KB, Danehy FT, Kavosi M, Kumar S, Lin S, Littlefield C, Looby K, Manohar R, Martin CJ, Wood M, Zawadzka A, Wawersik S, Nicholls SB, Datta A, Buckler A, Schürpf T, Carven GJ, Qatanani M, Fogel AI. An antibody that inhibits TGF-β1 release from latent extracellular matrix complexes attenuates the progression of renal fibrosis. Sci Signal 2024; 17:eadn6052. [PMID: 38980922 DOI: 10.1126/scisignal.adn6052] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2024] [Accepted: 06/11/2024] [Indexed: 07/11/2024]
Abstract
Inhibitors of the transforming growth factor-β (TGF-β) pathway are potentially promising antifibrotic therapies, but nonselective simultaneous inhibition of all three TGF-β homologs has safety liabilities. TGF-β1 is noncovalently bound to a latency-associated peptide that is, in turn, covalently bound to different presenting molecules within large latent complexes. The latent TGF-β-binding proteins (LTBPs) present TGF-β1 in the extracellular matrix, and TGF-β1 is presented on immune cells by two transmembrane proteins, glycoprotein A repetitions predominant (GARP) and leucine-rich repeat protein 33 (LRRC33). Here, we describe LTBP-49247, an antibody that selectively bound to and inhibited the activation of TGF-β1 presented by LTBPs but did not bind to TGF-β1 presented by GARP or LRRC33. Structural studies demonstrated that LTBP-49247 recognized an epitope on LTBP-presented TGF-β1 that is not accessible on GARP- or LRRC33-presented TGF-β1, explaining the antibody's selectivity for LTBP-complexed TGF-β1. In two rodent models of kidney fibrosis of different etiologies, LTBP-49247 attenuated fibrotic progression, indicating the central role of LTBP-presented TGF-β1 in renal fibrosis. In mice, LTBP-49247 did not have the toxic effects associated with less selective TGF-β inhibitors. These results establish the feasibility of selectively targeting LTBP-bound TGF-β1 as an approach for treating fibrosis.
Collapse
Affiliation(s)
| | | | - Ajai Pal
- Scholar Rock Inc., 301 Binney Street, Cambridge, MA 02142, USA
| | - George Coricor
- Scholar Rock Inc., 301 Binney Street, Cambridge, MA 02142, USA
| | - Chris Boston
- Scholar Rock Inc., 301 Binney Street, Cambridge, MA 02142, USA
| | | | | | | | - Shaun Cote
- Scholar Rock Inc., 301 Binney Street, Cambridge, MA 02142, USA
| | - Kevin B Dagbay
- Scholar Rock Inc., 301 Binney Street, Cambridge, MA 02142, USA
| | | | - Mania Kavosi
- Scholar Rock Inc., 301 Binney Street, Cambridge, MA 02142, USA
| | - Sandeep Kumar
- Scholar Rock Inc., 301 Binney Street, Cambridge, MA 02142, USA
| | - Susan Lin
- Scholar Rock Inc., 301 Binney Street, Cambridge, MA 02142, USA
| | | | - Kailyn Looby
- Scholar Rock Inc., 301 Binney Street, Cambridge, MA 02142, USA
| | - Rohan Manohar
- Scholar Rock Inc., 301 Binney Street, Cambridge, MA 02142, USA
| | | | - Marcie Wood
- Scholar Rock Inc., 301 Binney Street, Cambridge, MA 02142, USA
- ToxStrategies LLC, 23501 Cinco Ranch Boulevard, Katy, TX 77494, USA
| | - Agatha Zawadzka
- Scholar Rock Inc., 301 Binney Street, Cambridge, MA 02142, USA
| | - Stefan Wawersik
- Scholar Rock Inc., 301 Binney Street, Cambridge, MA 02142, USA
| | | | - Abhishek Datta
- Scholar Rock Inc., 301 Binney Street, Cambridge, MA 02142, USA
| | - Alan Buckler
- Scholar Rock Inc., 301 Binney Street, Cambridge, MA 02142, USA
| | - Thomas Schürpf
- Scholar Rock Inc., 301 Binney Street, Cambridge, MA 02142, USA
| | | | | | - Adam I Fogel
- Scholar Rock Inc., 301 Binney Street, Cambridge, MA 02142, USA
| |
Collapse
|
2
|
Kossatz S, Beer AJ, Notni J. It's Time to Shift the Paradigm: Translation and Clinical Application of Non-αvβ3 Integrin Targeting Radiopharmaceuticals. Cancers (Basel) 2021; 13:cancers13235958. [PMID: 34885066 PMCID: PMC8657165 DOI: 10.3390/cancers13235958] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2021] [Revised: 11/16/2021] [Accepted: 11/17/2021] [Indexed: 11/16/2022] Open
Abstract
Simple Summary Cancer cells often present a different set of proteins on their surface than normal cells. This also applies to integrins, a class of 24 cell surface receptors which mainly are responsible for physically anchoring cells in tissues, but also fulfil a plethora of other functions. If a certain integrin is found on tumor cells but not on normal ones, radioactive molecules (named tracers) that specifically bind to this integrin will accumulate in the cancer lesion if injected into the blood stream. The emitted radiation can be detected from outside the body and allows for localization and thus, diagnosis, of cancer. Only one of the 24 integrins, the subtype αvβ3, has hitherto been thoroughly investigated in this context. We herein summarize the most recent, pertinent research on other integrins, and argue that some of these approaches might ultimately improve the clinical management of the most lethal cancers, such as pancreatic carcinoma. Abstract For almost the entire period of the last two decades, translational research in the area of integrin-targeting radiopharmaceuticals was strongly focused on the subtype αvβ3, owing to its expression on endothelial cells and its well-established role as a biomarker for, and promoter of, angiogenesis. Despite a large number of translated tracers and clinical studies, a clinical value of αvβ3-integrin imaging could not be defined yet. The focus of research has, thus, been moving slowly but steadily towards other integrin subtypes which are involved in a large variety of tumorigenic pathways. Peptidic and non-peptidic radioligands for the integrins α5β1, αvβ6, αvβ8, α6β1, α6β4, α3β1, α4β1, and αMβ2 were first synthesized and characterized preclinically. Some of these compounds, targeting the subtypes αvβ6, αvβ8, and α6β1/β4, were subsequently translated into humans during the last few years. αvβ6-Integrin has arguably attracted most attention because it is expressed by some of the cancers with the worst prognosis (above all, pancreatic ductal adenocarcinoma), which substantiates a clinical need for the respective theranostic agents. The receptor furthermore represents a biomarker for malignancy and invasiveness of carcinomas, as well as for fibrotic diseases, such as idiopathic pulmonary fibrosis (IPF), and probably even for Sars-CoV-2 (COVID-19) related syndromes. Accordingly, the largest number of recent first-in-human applications has been reported for radiolabeled compounds targeting αvβ6-integrin. The results indicate a substantial clinical value, which might lead to a paradigm change and trigger the replacement of αvβ3 by αvβ6 as the most popular integrin in theranostics.
Collapse
Affiliation(s)
- Susanne Kossatz
- Department of Nuclear Medicine, School of Medicine, Technical University of Munich, 81675 Munich, Germany;
- Central Institute for Translational Cancer Research (TranslaTUM), School of Medicine, Technical University of Munich, 81675 Munich, Germany
| | | | - Johannes Notni
- Department of Pathology, School of Medicine, Technical University of Munich, 81675 Munich, Germany
- TRIMT GmbH, 01454 Radeberg, Germany
- Correspondence: ; Tel.: +49-89-4140-6075; Fax: +49-89-4140-6949
| |
Collapse
|
3
|
Seed RI, Kobayashi K, Ito S, Takasaka N, Cormier A, Jespersen JM, Publicover J, Trilok S, Combes AJ, Chew NW, Chapman J, Krummel MF, Lou J, Marks J, Cheng Y, Baron JL, Nishimura SL. A tumor-specific mechanism of T reg enrichment mediated by the integrin αvβ8. Sci Immunol 2021; 6:6/57/eabf0558. [PMID: 33771888 DOI: 10.1126/sciimmunol.abf0558] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2020] [Revised: 01/06/2021] [Accepted: 02/24/2021] [Indexed: 12/14/2022]
Abstract
Regulatory T cells (Tregs) that promote tumor immune evasion are enriched in certain tumors and correlate with poor prognosis. However, mechanisms for Treg enrichment remain incompletely understood. We described a mechanism for Treg enrichment in mouse and human tumors mediated by the αvβ8 integrin. Tumor cell αvβ8 bound to latent transforming growth factor-β (L-TGF-β) presented on the surface of T cells, resulting in TGF-β activation and immunosuppressive Treg differentiation in vitro. In vivo, tumor cell αvβ8 expression correlated with Treg enrichment, immunosuppressive Treg gene expression, and increased tumor growth, which was reduced in mice by αvβ8 inhibition or Treg depletion. Structural modeling and cell-based studies suggested a highly geometrically constrained complex forming between αvβ8-expressing tumor cells and L-TGF-β-expressing T cells, facilitating TGF-β activation, independent of release and diffusion, and providing limited access to TGF-β inhibitors. These findings suggest a highly localized tumor-specific mechanism for Treg enrichment.
Collapse
Affiliation(s)
- Robert I Seed
- Department of Pathology, University of California, San Francisco, San Francisco, CA 94110, USA
| | - Kenji Kobayashi
- Department of Pathology, University of California, San Francisco, San Francisco, CA 94110, USA
| | - Saburo Ito
- Department of Pathology, University of California, San Francisco, San Francisco, CA 94110, USA
| | - Naoki Takasaka
- Department of Pathology, University of California, San Francisco, San Francisco, CA 94110, USA
| | - Anthony Cormier
- Department of Pathology, University of California, San Francisco, San Francisco, CA 94110, USA
| | - Jillian M Jespersen
- Department of Medicine and Liver Center, University of California, San Francisco, San Francisco, CA 94143, USA
| | - Jean Publicover
- Department of Medicine and Liver Center, University of California, San Francisco, San Francisco, CA 94143, USA
| | - Suprita Trilok
- Department of Medicine and Liver Center, University of California, San Francisco, San Francisco, CA 94143, USA
| | - Alexis J Combes
- Department of Pathology, University of California, San Francisco, San Francisco, CA 94110, USA.,ImmunoX Initiative, University of California, San Francisco, San Francisco, CA 94143, USA.,ImmunoX CoLabs, University of California San Francisco, San Francisco, CA 94143, USA
| | - Nayvin W Chew
- Department of Pathology, University of California, San Francisco, San Francisco, CA 94110, USA.,ImmunoX Initiative, University of California, San Francisco, San Francisco, CA 94143, USA.,ImmunoX CoLabs, University of California San Francisco, San Francisco, CA 94143, USA
| | - Jocelyne Chapman
- Department of Gynecology and Oncology, University of California, San Francisco San Francisco, CA 94110, USA
| | - Matthew F Krummel
- Department of Pathology, University of California, San Francisco, San Francisco, CA 94110, USA.,ImmunoX Initiative, University of California, San Francisco, San Francisco, CA 94143, USA
| | - Jianlong Lou
- Department of Anesthesia and Perioperative Care, University of California, San Francisco, San Francisco, CA 94110, USA
| | - James Marks
- Department of Anesthesia and Perioperative Care, University of California, San Francisco, San Francisco, CA 94110, USA
| | - Yifan Cheng
- Department of Biochemistry and Biophysics, University of California, San Francisco, San Francisco, CA 94158, USA.,Howard Hughes Medical Institute, University of California, San Francisco, San Francisco, CA 94158, USA
| | - Jody L Baron
- Department of Medicine and Liver Center, University of California, San Francisco, San Francisco, CA 94143, USA.,ImmunoX Initiative, University of California, San Francisco, San Francisco, CA 94143, USA
| | - Stephen L Nishimura
- Department of Pathology, University of California, San Francisco, San Francisco, CA 94110, USA. .,ImmunoX Initiative, University of California, San Francisco, San Francisco, CA 94143, USA
| |
Collapse
|
4
|
Welsh BT, Faucette R, Bilic S, Martin CJ, Schürpf T, Chen D, Nicholls S, Lansita J, Kalra A. Nonclinical Development of SRK-181: An Anti-Latent TGFβ1 Monoclonal Antibody for the Treatment of Locally Advanced or Metastatic Solid Tumors. Int J Toxicol 2021; 40:226-241. [PMID: 33739172 PMCID: PMC8135237 DOI: 10.1177/1091581821998945] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
Checkpoint inhibitors offer a promising immunotherapy strategy for cancer treatment; however, due to primary or acquired resistance, many patients do not achieve lasting clinical responses. Recently, the transforming growth factor-β (TGFβ) signaling pathway has been identified as a potential target to overcome primary resistance, although the nonselective inhibition of multiple TGFβ isoforms has led to dose-limiting cardiotoxicities. SRK-181 is a high-affinity, fully human antibody that selectively binds to latent TGFβ1 and inhibits its activation. To support SRK-181 clinical development, we present here a comprehensive preclinical assessment of its pharmacology, pharmacokinetics, and safety across multiple species. In vitro studies showed that SRK-181 has no effect on human platelet function and does not induce cytokine release in human peripheral blood. Four-week toxicology studies with SRK-181 showed that weekly intravenous administration achieved sustained serum exposure and was well tolerated in rats and monkeys, with no treatment-related adverse findings. The no-observed-adverse-effect levels levels were 200 mg/kg in rats and 300 mg/kg in monkeys, the highest doses tested, and provide a nonclinical safety factor of up to 813-fold (based on Cmax) above the phase 1 starting dose of 80 mg every 3 weeks. In summary, the nonclinical pharmacology, pharmacokinetic, and toxicology data demonstrate that SRK-181 is a selective inhibitor of latent TGFβ1 that does not produce the nonclinical toxicities associated with nonselective TGFβ inhibition. These data support the initiation and safe conduct of a phase 1 trial with SRK-181 in patients with advanced cancer.
Collapse
Affiliation(s)
- Brian T Welsh
- 436132ToxStrategies, Research Blvd Building, Austin, TX, USA
| | | | | | | | | | | | | | | | | |
Collapse
|
5
|
Hall AP, Cauvin A, Dudal S, Raymond J, Rogerson P, Jolette J. Case Studies Discussing the Pathology, Immunogenicity, and Proposed Mechanism of Toxicity of an Inhaled Anti-TGFβ Humanized Fab Antibody in Non-Human Primates and Mice. Toxicol Pathol 2020; 49:315-333. [PMID: 33167807 DOI: 10.1177/0192623320960023] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022]
Abstract
Treatment of nonhuman primates and mice with a humanized antigen-binding fragment (Fab) antibody (UCBFab) inhibiting transforming growth factor β via daily inhalation for up to 13 weeks resulted in low systemic exposure but high local exposure in the lung. Target engagement was demonstrated by reduced levels of signal transducers, phosphoSMAD and plasminogen activator inhibitor-1 in the bronchoalveolar lavage fluid (BALF). Treatment was associated with a high frequency and titer of antidrug antibodies, indicating high local immunogenicity, and local pathology within the lung and draining lymph nodes. Microscopic changes were characterized by perivascular (PV) and peribronchiolar (PB) mononuclear inflammatory cell (MIC) infiltrates that were principally lymphocytic in nature and mixed inflammatory cell infiltrates and/or inflammation within the alveoli. Immunohistochemical investigation revealed a predominantly CD68-positive macrophage and CD3- and CD8>CD4-positive T-cell response in the alveoli, whereas within the airways, there was a variable mixture of CD3-positive T cells, CD20-positive B cells, and CD68-positive macrophages. Increased cellularity of the draining lymph nodes was also noted, indicating the presence of an immune response to the inhaled test article. Morphologic changes did not progress over time, and all changes partially recovered. Increased leukocytes (principally macrophages) in BALF cytology correlated with the changes seen by histopathology.
Collapse
Affiliation(s)
| | | | | | - James Raymond
- Charles River Pathology Associates, Frederick, MA, USA
| | | | | |
Collapse
|
6
|
Quigley NG, Steiger K, Richter F, Weichert W, Hoberück S, Kotzerke J, Notni J. Tracking a TGF-β activator in vivo: sensitive PET imaging of αvβ8-integrin with the Ga-68-labeled cyclic RGD octapeptide trimer Ga-68-Triveoctin. EJNMMI Res 2020; 10:133. [PMID: 33128636 PMCID: PMC7603442 DOI: 10.1186/s13550-020-00706-1] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2020] [Accepted: 09/21/2020] [Indexed: 01/02/2023] Open
Abstract
Purpose As a major activator of transforming growth factor β (TGF-β), the RGD receptor αvβ8-integrin is involved in pathogenic processes related to TGF-β dysregulation, such as tumor growth, invasion, and radiochemoresistance, metastasis and tumor cell stemness, as well as epithelial-mesenchymal transition. The novel positron emission tomography (PET) radiopharmaceutical Ga-68-Triveoctin for in vivo mapping of αvβ8-integrin expression might enhance the prognosis of certain tumor entities, as well as support and augment TGF-β-targeted therapeutic approaches. Methods Monomeric and trimeric conjugates of cyclo(GLRGDLp(NMe)K(pent-4-ynoic amide)) were synthesized by click chemistry (CuAAC), labeled with Ga-68, and evaluated in MeWo (human melanoma) xenografted SCID mice by means of PET and ex-vivo biodistribution. αvβ8-integrin expression in murine tissues was determined by β8-IHC. A human subject received a single injection of 173 MBq of Ga-68-Triveoctin and underwent 3 subsequent PET/CT scans at 25, 45, and 90 min p.i.. Results The trimer Ga-68-Triveoctin exhibits a 6.7-fold higher αvβ8-integrin affinity than the monomer (IC50 of 5.7 vs. 38 nM, respectively). Accordingly, biodistribution showed a higher tumor uptake (1.9 vs. 1.0%IA/g, respectively) but a similar baseline upon blockade (0.25%IA/g for both). IHC showed an intermediate β8-expression in the tumor while most organs and tissues were found β8-negative. Low non-target tissue uptakes (< 0.4%IA/g) confirmed a low degree of unspecific binding. Due to its hydrophilicity (log D = − 3.1), Ga-68-Triveoctin is excreted renally and shows favorable tumor/tissue ratios in mice (t/blood: 6.7; t/liver: 6.8; t/muscle: 29). A high kidney uptake in mice (kidney-to-blood and -to-muscle ratios of 126 and 505, respectively) is not reflected by human PET (corresponding values are 15 and 30, respectively), which furthermore showed notable uptakes in coeliac and choroid plexus (SUVmean 6.1 and 9.7, respectively, 90 min p.i.). Conclusion Ga-68-Triveoctin enables sensitive in-vivo imaging αvβ8-integrin expression in murine tumor xenografts. PET in a human subject confirmed a favorable biodistribution, underscoring the potential of Ga-68-Triveoctin for mapping of αvβ8-integrin expression in a clinical setting.
Collapse
Affiliation(s)
- Neil Gerard Quigley
- Institut für Pathologie Und Pathologische Anatomie, Technische Universität München, Trogerstraße 18, 81675, Munich, Germany
| | - Katja Steiger
- Institut für Pathologie Und Pathologische Anatomie, Technische Universität München, Trogerstraße 18, 81675, Munich, Germany
| | - Frauke Richter
- Institut für Pathologie Und Pathologische Anatomie, Technische Universität München, Trogerstraße 18, 81675, Munich, Germany
| | - Wilko Weichert
- Institut für Pathologie Und Pathologische Anatomie, Technische Universität München, Trogerstraße 18, 81675, Munich, Germany
| | - Sebastian Hoberück
- Klinik Und Poliklinik für Nuklearmedizin, Universitätsklinikum Carl Gustav Carus an der Technischen Universität Dresden, Dresden, Germany
| | - Jörg Kotzerke
- Klinik Und Poliklinik für Nuklearmedizin, Universitätsklinikum Carl Gustav Carus an der Technischen Universität Dresden, Dresden, Germany
| | - Johannes Notni
- Institut für Pathologie Und Pathologische Anatomie, Technische Universität München, Trogerstraße 18, 81675, Munich, Germany.
| |
Collapse
|
7
|
An In Vitro Model of Mast Cell Recruitment and Activation by Breast Cancer Cells Supports Anti-Tumoral Responses. Int J Mol Sci 2020; 21:ijms21155293. [PMID: 32722549 PMCID: PMC7432939 DOI: 10.3390/ijms21155293] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2020] [Revised: 05/29/2020] [Accepted: 06/18/2020] [Indexed: 12/18/2022] Open
Abstract
Breast cancer (BrC) affects millions of women yearly. Mast cells (MCs) are common components of breast tumors with documented agonistic and antagonistic roles in tumor progression. Understanding the participation of MCs in BrC may lead to new therapies to control tumor growth. In this study, we looked into mechanistic models of MC responses triggered by BrC cells (BrCC), assessing both early degranulation and late transcriptional activities. We used aggressive and non-aggressive BrCC to model the progressive staging of the disease over HMC1 and LAD-2 human MC lines. We found that both MC lines were chemoattracted by all BrCC, but their activation was preferentially induced by aggressive lines, finding differences in their active transcriptional programs, both at basal level and after stimulation. Among those genes with altered expression were down-regulated SPP1, PDCD1, IL17A and TGFB1 and up-regulated KITLG and IFNG. A low expression of SPP1 and a high expression of KITLG and IFNG were associated with increased overall survival of BrC patients from public databases. The set of altered genes is more often associated with tumor stromas enriched with anti-tumoral signals, suggesting that MCs may participate in tumor control.
Collapse
|
8
|
Abstract
Endoglin, known to be expressed in proliferating vessels, is of worth when evaluating microvessel density as a prognostic factor in many types of malignancies, including some subtypes of leukemia cells. In childhood acute lymphoblastic leukemia, endoglin is associated with adverse outcome. In bone marrow, endoglin identifies the repopulating hematopoietic stem cells. Mast cells are a component of normal tissue and play an important role in the regulation of several processes, including inflammation and neoplasia. The aim of this study was to evaluate the use of endoglin as a biological marker of mast cells compared with the gold standard stains. We studied 15 specimens of neurofibroma, 9 of mastocytosis, and 6 of fibrous scar tissue through immunohistochemistry (for endoglin and mast cell tryptase) and histochemical staining using toluidine blue. Quantitative analysis of the cells was performed by counting 5 hotspots. The validity of endoglin as a mast cell marker was assessed by intraclass correlation coefficient. The Kruskal-Wallis test was used to compare mast cell count for each marker. A strong endoglin expression was found in the cytoplasmic granules of mast cells within the 3 groups. Similar results were observed with mast cell tryptase as well as toluidine blue. The intraclass correlation coefficient revealed that endoglin is a highly reliable biomarker of mast cells when compared with mast cell tryptase and toluidine blue. In conclusion, endoglin may assist in the diagnosis and pathogenesis study of various processes associated with mast cells. An endoglin-neutralizing treatment for solid cancers and leukemia could also affect mastocytes and the immunologic system.
Collapse
|
9
|
Lyons DO, Pullen NA. Beyond IgE: Alternative Mast Cell Activation Across Different Disease States. Int J Mol Sci 2020; 21:ijms21041498. [PMID: 32098318 PMCID: PMC7073060 DOI: 10.3390/ijms21041498] [Citation(s) in RCA: 55] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2020] [Revised: 02/20/2020] [Accepted: 02/21/2020] [Indexed: 12/14/2022] Open
Abstract
Mast cells are often regarded through the lens of IgE-dependent reactions as a cell specialized only for anti-parasitic and type I hypersensitive responses. However, recently many researchers have begun to appreciate the expansive repertoire of stimuli that mast cells can respond to. After the characterization of the interleukin (IL)-33/suppression of tumorigenicity 2 (ST2) axis of mast cell activation-a pathway that is independent of the adaptive immune system-researchers are revisiting other stimuli to induce mast cell activation and/or subsequent degranulation independent of IgE. This discovery also underscores that mast cells act as important mediators in maintaining body wide homeostasis, especially through barrier defense, and can thus be the source of disease as well. Particularly in the gut, inflammatory bowel diseases (Crohn's disease, ulcerative colitis, etc.) are characterized with enhanced mast cell activity in the context of autoimmune disease. Mast cells show phenotypic differences based on tissue residency, which could manifest as different receptor expression profiles, allowing for unique mast cell responses (both IgE and non-IgE mediated) across varying tissues as well. This variety in receptor expression suggests mast cells respond differently, such as in the gut where immunosuppressive IL-10 stimulates the development of food allergy or in the lungs where transforming growth factor-β1 (TGF-β1) can enhance mast cell IL-6 production. Such differences in receptor expression illustrate the truly diverse effector capabilities of mast cells, and careful consideration must be given toward the phenotype of mast cells observed in vitro. Given mast cells' ubiquitous tissue presence and their capability to respond to a broad spectrum of non-IgE stimuli, it is expected that mast cells may also contribute to the progression of autoimmune disorders and other disease states such as metastatic cancer through promoting chronic inflammation in the local tissue microenvironment and ultimately polarizing toward a unique Th17 immune response. Furthermore, these interconnected, atypical activation pathways may crosstalk with IgE-mediated signaling differently across disorders such as parasitism, food allergies, and autoimmune disorders of the gut. In this review, we summarize recent research into familiar and novel pathways of mast cells activation and draw connections to clinical human disease.
Collapse
|
10
|
Ion homeostasis and transport are regulated by genes differentially expressed in porcine buccal pouch mucosal cells during long-term culture in vitro – a microarray approach. ACTA ACUST UNITED AC 2018. [DOI: 10.2478/acb-2018-0013] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/22/2023]
Abstract
Abstract
The oral mucosa is a compound tissue composed of several cells types, including fibroblasts and keratinocytes, that are characterized by different morphology, as well as biochemical and metabolomic properties. The oral mucosal cells are the most important factors mediated between transport and drugs delivery. The changes in cellular ion homeostasis may significantly affect the bioavailability of administrated drugs and their transport across the mucous membrane. Therefore we investigated the expression profile of genes involved in ion transport and homeostasis in porcine buccal pouch mucosal cells.
The oral mucosa was separated surgically and isolated enzymatically. The cells were examined during long-term in vitro culture (IVC). The cultured cells were collected at 7, 15 and 30 days of IVC and subsequently transferred to RNA isolation and next, the gene expression profile was measured using Affymetrix microarray assays.
In the results, we can extract genes belonging to four ontology groups: “ion homeostasis”, “ion transport”, “metal ion transport”, and “inorganic ion homeostasis”. For TGFB1 and CCL2, we observed up-regulation after 7 days of IVC, down-regulation after 15 days of IVC and upregulation again after 30 days of IVC. The ATP13A3, ATP1B1, CCL8, LYN, STEAP1, PDPN, PTGS2, and SLC5A3genes showed high activity after day 7 of IVC, and in the days 15 and 30 of IVC showed low activity.
We showed an expression profile of genes associated with the effects of ion influence on the porcine normal oral mucosal cell development in IVC. These studies may be the starting point for further research into oral diseases and will allow for the comparison of the gene expression profile of normal and disease altered cells.
Collapse
|
11
|
Lyons DO, Plewes MR, Pullen NA. Soluble transforming growth factor beta-1 enhances murine mast cell release of Interleukin 6 in IgE-independent and Interleukin 13 in IgE-dependent settings in vitro. PLoS One 2018; 13:e0207704. [PMID: 30444930 PMCID: PMC6239331 DOI: 10.1371/journal.pone.0207704] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2018] [Accepted: 11/03/2018] [Indexed: 12/05/2022] Open
Abstract
INTRODUCTION For immune cells transforming growth factor beta-1 (TGF-β1) can enhance or repress effector functions. Here, we characterize the effects of TGF-β1 on IgE-mediated and IL-33-mediated activation of primary murine mast cells derived from hematopoietic stem cells (bone marrow derived mast cells; BMMC). We also investigated potential interactions between TGF-β1 and stem cell factor (SCF). We conclude TGF-β1 plays a selectively stimulatory role for mast cell cultures in vitro. METHODS BMMCs from C57BL/6 mice were differentiated with IL-3 and then treated with TGF-β1. BMMCs were exposed to TGF-β1, primed with IgE, activated with antigen, and then IL-6 and IL-13 cytokine release was quantified using ELISA. Additionally, the effects of TGF-β1 on both IgE and IL-33-mediated short term activation were observed via flow cytometric analysis of both surface LAMP-1 expression and intracellular IL-6. Receptor colocalization was visualized using fluorescence confocal microscopy and individual receptor expression levels were also quantified. RESULTS Resting IL-6 production increased with TGF-β1 but significance was lost following BMMC activation via IgE receptor (FcεRI) crosslinking. This was similar to a comparison effect due to SCF treatment alone, which also enhanced resting levels of IL-6. TGF-β1 treatment enhanced release of IL-13 only with FcεRI-IgE-mediated activation. TGF-β1 suppressed mobilization of IL-6 with short-term BMMC activation when stimulated with IL-33. Lastly, colocalization patterns of the SCF receptor (CD117) and FcεRI with IgE crosslinking were unaffected by TGF-β1 treatment, but individual expression levels for FcεRI, CD117, and TGFβRII were all reduced following either IgE activation or TGF-β1 treatment; this reduction was partially recovered in BMMCs that were both activated by IgE and treated with TGF-β1. DISCUSSION These data reveal a novel positive effect of soluble TGF-β1 on mast cell activation in vitro, suggesting mast cells may be activated through a non-canonical pathway by TGF-β1. Understanding this interaction will provide insight into the potential role of mast cells in settings where TGF-β1 is produced in an aberrant manner, such as in and around high grade tumors.
Collapse
Affiliation(s)
- David O. Lyons
- School of Biological Sciences, University of Northern Colorado, Greeley, Colorado, United States of America
| | - Michele R. Plewes
- Olson Center for Women's Health, Department of Obstetrics and Gynecology, University of Nebraska Medical Center, Omaha, Nebraska, United States of America
| | - Nicholas A. Pullen
- School of Biological Sciences, University of Northern Colorado, Greeley, Colorado, United States of America
| |
Collapse
|
12
|
Takasaka N, Seed RI, Cormier A, Bondesson AJ, Lou J, Elattma A, Ito S, Yanagisawa H, Hashimoto M, Ma R, Levine MD, Publicover J, Potts R, Jespersen JM, Campbell MG, Conrad F, Marks JD, Cheng Y, Baron JL, Nishimura SL. Integrin αvβ8-expressing tumor cells evade host immunity by regulating TGF-β activation in immune cells. JCI Insight 2018; 3:122591. [PMID: 30333313 DOI: 10.1172/jci.insight.122591] [Citation(s) in RCA: 86] [Impact Index Per Article: 12.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2018] [Accepted: 08/30/2018] [Indexed: 12/27/2022] Open
Abstract
TGF-β is a promising immunotherapeutic target. It is expressed ubiquitously in a latent form that must be activated to function. Determination of where and how latent TGF-β (L-TGF-β) is activated in the tumor microenvironment could facilitate cell- and mechanism-specific approaches to immunotherapeutically target TGF-β. Binding of L-TGF-β to integrin αvβ8 results in activation of TGF-β. We engineered and used αvβ8 antibodies optimized for blocking or detection, which - respectively - inhibit tumor growth in syngeneic tumor models or sensitively and specifically detect β8 in human tumors. Inhibition of αvβ8 potentiates cytotoxic T cell responses and recruitment of immune cells to tumor centers - effects that are independent of PD-1/PD-L1. β8 is expressed on the cell surface at high levels by tumor cells, not immune cells, while the reverse is true of L-TGF-β, suggesting that tumor cell αvβ8 serves as a platform for activating cell-surface L-TGF-β presented by immune cells. Transcriptome analysis of tumor-associated lymphoid cells reveals macrophages as a key cell type responsive to β8 inhibition with major increases in chemokine and tumor-eliminating genes. High β8 expression in tumor cells is seen in 20%-80% of various cancers, which rarely coincides with high PD-L1 expression. These data suggest tumor cell αvβ8 is a PD-1/PD-L1-independent immunotherapeutic target.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | - Yifan Cheng
- Department of Biochemistry and Biophysics, and.,Howard Hughes Medical Institute, UCSF, San Francisco, California, USA
| | | | | |
Collapse
|
13
|
Campos JC, Cunha JD, Ferreira DC, Reis S, Costa PJ. Challenges in the local delivery of peptides and proteins for oral mucositis management. Eur J Pharm Biopharm 2018; 128:131-146. [PMID: 29702221 DOI: 10.1016/j.ejpb.2018.04.025] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2018] [Revised: 04/21/2018] [Accepted: 04/23/2018] [Indexed: 12/20/2022]
Abstract
Oral mucositis, a common inflammatory side effect of oncological treatments, is a disorder of the oral mucosa that can cause painful ulcerations, local motor disabilities, and an increased risk of infections. Due to the discomfort it produces and the associated health risks, it can lead to cancer treatment restrains, such as the need for dose reduction, cycle delays or abandonment. Current mucositis management has low efficiency in prevention and treatment. A topical drug application for a local action can be a more effective approach than systemic routes when addressing oral cavity pathologies. Local delivery of growth factors, antibodies, and anti-inflammatory cytokines have shown promising results. However, due to the peptide and protein nature of these novel agents, and the several anatomic, physiological and environmental challenges of the oral cavity, their local action might be limited when using traditional delivering systems. This review is an awareness of the issues and strategies in the local delivery of macromolecules for the management of oral mucositis.
Collapse
Affiliation(s)
- João C Campos
- UCIBIO, REQUIMTE, Laboratory of Pharmaceutical Technology, Department of Drug Sciences, Faculty of Pharmacy, University of Porto, Portugal(1).
| | - João D Cunha
- UCIBIO, REQUIMTE, Laboratory of Pharmaceutical Technology, Department of Drug Sciences, Faculty of Pharmacy, University of Porto, Portugal(1)
| | - Domingos C Ferreira
- UCIBIO, REQUIMTE, Laboratory of Pharmaceutical Technology, Department of Drug Sciences, Faculty of Pharmacy, University of Porto, Portugal(1)
| | - Salette Reis
- LAQV, REQUIMTE, Department of Chemical Sciences, Faculty of Pharmacy, University of Porto, Portugal(1)
| | - Paulo J Costa
- UCIBIO, REQUIMTE, Laboratory of Pharmaceutical Technology, Department of Drug Sciences, Faculty of Pharmacy, University of Porto, Portugal(1)
| |
Collapse
|
14
|
Comparison study between plasma rich in growth factors and platelet-rich plasma for osteoconduction in rat calvaria. JOURNAL OF ORAL AND MAXILLOFACIAL SURGERY MEDICINE AND PATHOLOGY 2017. [DOI: 10.1016/j.ajoms.2017.06.011] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/13/2023]
|
15
|
Yanagisawa H, Hashimoto M, Minagawa S, Takasaka N, Ma R, Moermans C, Ito S, Araya J, Budelsky A, Goodsell A, Baron JL, Nishimura SL. Role of IL-17A in murine models of COPD airway disease. Am J Physiol Lung Cell Mol Physiol 2016; 312:L122-L130. [PMID: 27913421 DOI: 10.1152/ajplung.00301.2016] [Citation(s) in RCA: 44] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2016] [Revised: 11/22/2016] [Accepted: 11/22/2016] [Indexed: 12/20/2022] Open
Abstract
Small airway fibrosis is a major pathological feature of chronic obstructive pulmonary disease (COPD) and is refractory to current treatments. Chronic inflammatory cells accumulate around small airways in COPD and are thought to play a major role in small airway fibrosis. Mice deficient in α/β T cells have recently been shown to be protected from both experimental airway inflammation and fibrosis. In these models, CD4+Th17 cells and secretion of IL-17A are increased. However, a pathogenic role for IL-17 in specifically mediating fibrosis around airways has not been demonstrated. Here a role for IL-17A in airway fibrosis was demonstrated using mice deficient in the IL-17 receptor A (il17ra) Il17ra-deficient mice were protected from both airway inflammation and fibrosis in two different models of airway fibrosis that employ COPD-relevant stimuli. In these models, CD4+ Th17 are a major source of IL-17A with other expressing cell types including γδ T cells, type 3 innate lymphoid cells, polymorphonuclear cells, and CD8+ T cells. Antibody neutralization of IL-17RA or IL-17A confirmed that IL-17A was the relevant pathogenic IL-17 isoform and IL-17RA was the relevant receptor in airway inflammation and fibrosis. These results demonstrate that the IL-17A/IL-17 RA axis is crucial to murine airway fibrosis. These findings suggest that IL-17 might be targeted to prevent the progression of airway fibrosis in COPD.
Collapse
Affiliation(s)
- Haruhiko Yanagisawa
- Department of Pathology, University of California, San Francisco, California
| | - Mitsuo Hashimoto
- Department of Pathology, University of California, San Francisco, California
| | - Shunsuke Minagawa
- Department of Pathology, University of California, San Francisco, California
| | - Naoki Takasaka
- Department of Pathology, University of California, San Francisco, California
| | - Royce Ma
- Department of Pathology, University of California, San Francisco, California
| | - Catherine Moermans
- Department of Pathology, University of California, San Francisco, California
| | - Saburo Ito
- Department of Pathology, University of California, San Francisco, California
| | - Jun Araya
- Department of Internal Medicine, Respiratory Division, Jikei University, Tokyo, Japan; and
| | - Alison Budelsky
- Department of Inflammation Research, Amgen, Seattle, Washington
| | - Amanda Goodsell
- Department of Medicine, University of California, San Francisco, California
| | - Jody L Baron
- Department of Medicine, University of California, San Francisco, California
| | - Stephen L Nishimura
- Department of Pathology, University of California, San Francisco, California;
| |
Collapse
|
16
|
Zhao H, Zhao L, Zhou Z, Wu Y. The roles of connective tissue growth factor in the development of anastomotic esophageal strictures. Arch Med Sci 2015; 11:770-8. [PMID: 26322089 PMCID: PMC4548024 DOI: 10.5114/aoms.2015.48147] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/16/2013] [Revised: 06/12/2013] [Accepted: 08/22/2013] [Indexed: 12/14/2022] Open
Abstract
INTRODUCTION The aim of this study was to investigate the roles of connective tissue growth factor (CTGF) in the development of anastomotic strictures after surgical repair of the esophagus. MATERIAL AND METHODS Tissues collected from the patients were divided into three groups based on the results of endoscopy and clinical grading. Patients without dysphagia after esophagectomy were used as the control population. The protein levels of CTGF, TGF-β1, Smad2, and Smad4 were determined by immunohistochemistry (IHC) and western blot analyses, while the mRNA levels of the two growth factors were evaluated by real-time polymerase chain reaction. RESULTS Compared with the control group, significantly increased (p < 0.01) levels of CTGF and TGF-β1 protein were observed in the anastomotic stenosis (AS) group, and levels of the two proteins detected by the IHC and western blot analyses were also significantly increased with the increasing severity of stenosis (p < 0.05). The mRNA levels of CTGF and TGF-β1 in the tissues collected from the patients with stenosis were significantly up-regulated (p < 0.05) as compared with those from the control group. In addition, the levels of Smad2 and Smad4 protein were also significantly increased (p < 0.05) with the increasing severity of stenosis, and the protein levels were positively correlated with the levels of CTGF (r = 0.59, p < 0.05) and TGF-β1 (r = 0.63, p < 0.05). CONCLUSIONS Inhibition of CTGF protein or mRNA expression may be a distinctive and effective therapy for the treatment of postoperative anastomotic strictures.
Collapse
Affiliation(s)
- Haibin Zhao
- Department of Pathology, the 101 Hospital of Chinese People's Liberation Army, Wuxi, Jiangsu Province, China
| | - Lingna Zhao
- Department of Pathology, the 101 Hospital of Chinese People's Liberation Army, Wuxi, Jiangsu Province, China
| | - Zhihua Zhou
- Department of Pathology, the 101 Hospital of Chinese People's Liberation Army, Wuxi, Jiangsu Province, China
| | - Yaoyi Wu
- Department of Pathology, the 101 Hospital of Chinese People's Liberation Army, Wuxi, Jiangsu Province, China
| |
Collapse
|
17
|
Woods LT, Camden JM, El-Sayed FG, Khalafalla MG, Petris MJ, Erb L, Weisman GA. Increased Expression of TGF-β Signaling Components in a Mouse Model of Fibrosis Induced by Submandibular Gland Duct Ligation. PLoS One 2015; 10:e0123641. [PMID: 25955532 PMCID: PMC4425516 DOI: 10.1371/journal.pone.0123641] [Citation(s) in RCA: 49] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2014] [Accepted: 02/21/2015] [Indexed: 02/06/2023] Open
Abstract
Transforming growth factor-β (TGF-β) is a multi-functional cytokine with a well-described role in the regulation of tissue fibrosis and regeneration in the liver, kidney and lung. Submandibular gland (SMG) duct ligation and subsequent deligation in rodents is a classical model for studying salivary gland damage and regeneration. While previous studies suggest that TGF-β may contribute to salivary gland fibrosis, the expression of TGF-β signaling components has not been investigated in relation to mouse SMG duct ligation-induced fibrosis and regeneration following ductal deligation. Following a 7 day SMG duct ligation, TGF-β1 and TGF-β3 were significantly upregulated in the SMG, as were TGF-β receptor 1 and downstream Smad family transcription factors in salivary acinar cells, but not in ductal cells. In acinar cells, duct ligation also led to upregulation of snail, a Smad-activated E-cadherin repressor and regulator of epithelial-mesenchymal transition, whereas in ductal cells upregulation of E-cadherin was observed while snail expression was unchanged. Upregulation of these TGF-β signaling components correlated with upregulation of fibrosis markers collagen 1 and fibronectin, responses that were inhibited by administration of the TGF-β receptor 1 inhibitors SB431542 or GW788388. After SMG regeneration following a 28 day duct deligation, TGF-β signaling components and epithelial-mesenchymal transition markers returned to levels similar to non-ligated controls. The results from this study indicate that increased TGF-β signaling contributes to duct ligation-induced changes in salivary epithelium that correlate with glandular fibrosis. Furthermore, the reversibility of enhanced TGF-β signaling in acinar cells of duct-ligated mouse SMG after deligation indicates that this is an ideal model for studying TGF-β signaling mechanisms in salivary epithelium as well as mechanisms of fibrosis initiation and their resolution.
Collapse
Affiliation(s)
- Lucas T. Woods
- Department of Biochemistry, University of Missouri, Columbia, Missouri, United States of America
- Christopher S. Bond Life Sciences Center, University of Missouri, Columbia, Missouri, United States of America
| | - Jean M. Camden
- Department of Biochemistry, University of Missouri, Columbia, Missouri, United States of America
- Christopher S. Bond Life Sciences Center, University of Missouri, Columbia, Missouri, United States of America
| | - Farid G. El-Sayed
- Department of Biochemistry, University of Missouri, Columbia, Missouri, United States of America
- Christopher S. Bond Life Sciences Center, University of Missouri, Columbia, Missouri, United States of America
| | - Mahmoud G. Khalafalla
- Department of Biochemistry, University of Missouri, Columbia, Missouri, United States of America
- Christopher S. Bond Life Sciences Center, University of Missouri, Columbia, Missouri, United States of America
| | - Michael J. Petris
- Department of Biochemistry, University of Missouri, Columbia, Missouri, United States of America
- Christopher S. Bond Life Sciences Center, University of Missouri, Columbia, Missouri, United States of America
- Department of Nutritional Sciences and Exercise Physiology, University of Missouri, Columbia, Missouri, United States of America
| | - Laurie Erb
- Department of Biochemistry, University of Missouri, Columbia, Missouri, United States of America
- Christopher S. Bond Life Sciences Center, University of Missouri, Columbia, Missouri, United States of America
| | - Gary A. Weisman
- Department of Biochemistry, University of Missouri, Columbia, Missouri, United States of America
- Christopher S. Bond Life Sciences Center, University of Missouri, Columbia, Missouri, United States of America
- * E-mail:
| |
Collapse
|
18
|
Brand OJ, Somanath S, Moermans C, Yanagisawa H, Hashimoto M, Cambier S, Markovics J, Bondesson AJ, Hill A, Jablons D, Wolters P, Lou J, Marks JD, Baron JL, Nishimura SL. Transforming Growth Factor-β and Interleukin-1β Signaling Pathways Converge on the Chemokine CCL20 Promoter. J Biol Chem 2015; 290:14717-28. [PMID: 25918170 DOI: 10.1074/jbc.m114.630368] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2014] [Indexed: 12/27/2022] Open
Abstract
CCL20 is the only chemokine ligand for the chemokine receptor CCR6, which is expressed by the critical antigen presenting cells, dendritic cells. Increased expression of CCL20 is likely involved in the increased recruitment of dendritic cells observed in fibroinflammatory diseases such as chronic obstructive pulmonary disease (COPD). CCL20 expression is increased by the proinflammatory cytokine IL-1β. We have determined that IL-1β-dependent CCL20 expression is also dependent on the multifunctional cytokine TGF-β. TGF-β is expressed in a latent form that must be activated to function, and activation is achieved through binding to the integrin αvβ8 (itgb8). Here we confirm correlative increases in αvβ8 and IL-1β with CCL20 protein in lung parenchymal lysates of a large cohort of COPD patients. How IL-1β- and αvβ8-mediated TGF-β activation conspire to increase fibroblast CCL20 expression remains unknown, because these pathways have not been shown to directly interact. We evaluate the 5'-flanking region of CCL20 to determine that IL-1β-driven CCL20 expression is dependent on αvβ8-mediated activation of TGF-β. We identify a TGF-β-responsive element (i.e. SMAD) located on an upstream enhancer of the human CCL20 promoter required for efficient IL-1β-dependent CCL20 expression. By chromatin immunoprecipitation, this upstream enhancer complexes with the p50 subunit of NF-κB on a NF-κB-binding element close to the transcriptional start site of CCL20. These interactions are confirmed by electromobility shift assays in nuclear extracts from human lung fibroblasts. These data define a mechanism by which αvβ8-dependent activation of TGF-β regulates IL-1β-dependent CCL20 expression in COPD.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | | | - Arthur Hill
- Surgery, University of California, San Francisco, California 94110
| | - David Jablons
- Surgery, University of California, San Francisco, California 94110
| | | | | | | | | | | |
Collapse
|
19
|
Minagawa S, Lou J, Seed RI, Cormier A, Wu S, Cheng Y, Murray L, Tsui P, Connor J, Herbst R, Govaerts C, Barker T, Cambier S, Yanagisawa H, Goodsell A, Hashimoto M, Brand OJ, Cheng R, Ma R, McKnelly KJ, Wen W, Hill A, Jablons D, Wolters P, Kitamura H, Araya J, Barczak AJ, Erle DJ, Reichardt LF, Marks JD, Baron JL, Nishimura SL. Selective targeting of TGF-β activation to treat fibroinflammatory airway disease. Sci Transl Med 2015; 6:241ra79. [PMID: 24944194 DOI: 10.1126/scitranslmed.3008074] [Citation(s) in RCA: 72] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Abstract
Airway remodeling, caused by inflammation and fibrosis, is a major component of chronic obstructive pulmonary disease (COPD) and currently has no effective treatment. Transforming growth factor-β (TGF-β) has been widely implicated in the pathogenesis of airway remodeling in COPD. TGF-β is expressed in a latent form that requires activation. The integrin αvβ8 (encoded by the itgb8 gene) is a receptor for latent TGF-β and is essential for its activation. Expression of integrin αvβ8 is increased in airway fibroblasts in COPD and thus is an attractive therapeutic target for the treatment of airway remodeling in COPD. We demonstrate that an engineered optimized antibody to human αvβ8 (B5) inhibited TGF-β activation in transgenic mice expressing only human and not mouse ITGB8. The B5 engineered antibody blocked fibroinflammatory responses induced by tobacco smoke, cytokines, and allergens by inhibiting TGF-β activation. To clarify the mechanism of action of B5, we used hydrodynamic, mutational, and electron microscopic methods to demonstrate that αvβ8 predominantly adopts a constitutively active, extended-closed headpiece conformation. Epitope mapping and functional characterization of B5 revealed an allosteric mechanism of action due to locking-in of a low-affinity αvβ8 conformation. Collectively, these data demonstrate a new model for integrin function and present a strategy to selectively target the TGF-β pathway to treat fibroinflammatory airway diseases.
Collapse
Affiliation(s)
- Shunsuke Minagawa
- Department of Pathology, University of California, San Francisco, San Francisco, CA 94110, USA
| | - Jianlong Lou
- Department of Anesthesia and Perioperative Care, University of California, San Francisco, San Francisco, CA 94110, USA
| | - Robert I Seed
- Department of Pathology, University of California, San Francisco, San Francisco, CA 94110, USA
| | - Anthony Cormier
- Department of Pathology, University of California, San Francisco, San Francisco, CA 94110, USA
| | - Shenping Wu
- The Keck Advanced Microscopy Laboratory, Department of Biochemistry and Biophysics, University of California, San Francisco, San Francisco, CA 94110, USA
| | - Yifan Cheng
- The Keck Advanced Microscopy Laboratory, Department of Biochemistry and Biophysics, University of California, San Francisco, San Francisco, CA 94110, USA
| | - Lynne Murray
- Department of Respiratory, Inflammation and Autoimmunity, MedImmune, Gaithersburg, MD 20878, USA. Department of Respiratory, Inflammation and Autoimmunity, MedImmune, Cambridge CB21 6GH, UK
| | - Ping Tsui
- Department of Respiratory, Inflammation and Autoimmunity, MedImmune, Gaithersburg, MD 20878, USA
| | - Jane Connor
- Department of Respiratory, Inflammation and Autoimmunity, MedImmune, Gaithersburg, MD 20878, USA
| | - Ronald Herbst
- Department of Respiratory, Inflammation and Autoimmunity, MedImmune, Gaithersburg, MD 20878, USA
| | - Cedric Govaerts
- Department of Respiratory, Inflammation and Autoimmunity, MedImmune, Cambridge CB21 6GH, UK
| | - Tyren Barker
- Department of Pathology, University of California, San Francisco, San Francisco, CA 94110, USA
| | - Stephanie Cambier
- Department of Pathology, University of California, San Francisco, San Francisco, CA 94110, USA
| | - Haruhiko Yanagisawa
- Department of Pathology, University of California, San Francisco, San Francisco, CA 94110, USA
| | - Amanda Goodsell
- Department of Medicine, University of California, San Francisco, San Francisco, CA 94110, USA
| | - Mitsuo Hashimoto
- Department of Pathology, University of California, San Francisco, San Francisco, CA 94110, USA
| | - Oliver J Brand
- Department of Pathology, University of California, San Francisco, San Francisco, CA 94110, USA
| | - Ran Cheng
- Department of Pathology, University of California, San Francisco, San Francisco, CA 94110, USA
| | - Royce Ma
- Department of Pathology, University of California, San Francisco, San Francisco, CA 94110, USA
| | - Kate J McKnelly
- Department of Pathology, University of California, San Francisco, San Francisco, CA 94110, USA
| | - Weihua Wen
- Department of Anesthesia and Perioperative Care, University of California, San Francisco, San Francisco, CA 94110, USA
| | - Arthur Hill
- Department of Surgery, University of California, San Francisco, San Francisco, CA 94110, USA
| | - David Jablons
- Department of Surgery, University of California, San Francisco, San Francisco, CA 94110, USA
| | - Paul Wolters
- Department of Medicine, University of California, San Francisco, San Francisco, CA 94110, USA
| | - Hideya Kitamura
- Department of Pathology, University of California, San Francisco, San Francisco, CA 94110, USA
| | - Jun Araya
- Department of Pulmonary Medicine, Jikei University, Tokyo 105 8461, Japan
| | - Andrea J Barczak
- Department of Medicine, University of California, San Francisco, San Francisco, CA 94110, USA
| | - David J Erle
- Department of Medicine, University of California, San Francisco, San Francisco, CA 94110, USA
| | - Louis F Reichardt
- Genetics, Development, and Behavioral Sciences, University of California, San Francisco, San Francisco, CA 94110, USA
| | - James D Marks
- Department of Anesthesia and Perioperative Care, University of California, San Francisco, San Francisco, CA 94110, USA
| | - Jody L Baron
- Department of Medicine, University of California, San Francisco, San Francisco, CA 94110, USA
| | - Stephen L Nishimura
- Department of Pathology, University of California, San Francisco, San Francisco, CA 94110, USA.
| |
Collapse
|
20
|
Cutaneous keratoacanthomas/squamous cell carcinomas associated with neutralization of transforming growth factor β by the monoclonal antibody fresolimumab (GC1008). Cancer Immunol Immunother 2015; 64:437-46. [PMID: 25579378 DOI: 10.1007/s00262-015-1653-0] [Citation(s) in RCA: 119] [Impact Index Per Article: 11.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/24/2013] [Accepted: 12/31/2014] [Indexed: 02/08/2023]
Abstract
Fresolimumab is an antibody capable of neutralizing all human isoforms of transforming growth factor beta (TGFβ) and has demonstrated anticancer activity in investigational studies. Inhibition of TGFβ by fresolimumab can potentially result in the development of cutaneous lesions. The aim of this study was to investigate the clinical, histological, and immunohistochemical characteristics of cutaneous neoplasms associated with fresolimumab. Skin biopsies (n = 24) were collected and analyzed from patients (n = 5) with treatment-emergent, cutaneous lesions arising during a phase 1 study of multiple doses of fresolimumab in patients (n = 29) with melanoma or renal cell carcinoma. Blinded, independent histological review and measurements of Ki-67, p53, and HPV integration were performed. Based on central review, four patients developed lesions with histological characteristics of keratoacanthomas, and of these patients, a single case of well-differentiated squamous cell carcinoma was also found. Expression of Ki-67, no evidence of p53 overexpression, and only focal positivity for human papillomavirus RNA by in situ hybridization in 4/18 cases were consistent with these findings. Following completion of fresolimumab, lesions spontaneously resolved. Therefore, benign, reversible keratoacanthomas were the most common cutaneous neoplasms observed, a finding of importance for adverse event monitoring, patient care, and optimization of therapies targeting TGFβ.
Collapse
|
21
|
Eda T, Takahashi K, Iwai S, Ogura N, Ito K, Tsukahara H, Suemitsu M, Kuboyama N, Kuyama K, Kondoh T. Effects of Plasma Rich in Growth Factors on Bone Formation in Rat Calvaria. J HARD TISSUE BIOL 2015. [DOI: 10.2485/jhtb.24.61] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/24/2022]
Affiliation(s)
- Takashi Eda
- Department of Maxillofacial Surgery, Nihon University School of Dentistry at Matsudo
| | - Kosuke Takahashi
- Research Institute of oral science, Nihon University School of Dentistry at Matsudo
- Department of Maxillofacial Surgery, Nihon University School of Dentistry at Matsudo
| | - Satoshi Iwai
- Research Institute of oral science, Nihon University School of Dentistry at Matsudo
- Department of Maxillofacial Surgery, Nihon University School of Dentistry at Matsudo
| | - Naomi Ogura
- Research Institute of oral science, Nihon University School of Dentistry at Matsudo
- Department of Maxillofacial Surgery, Nihon University School of Dentistry at Matsudo
| | - Ko Ito
- Research Institute of oral science, Nihon University School of Dentistry at Matsudo
- Department of Maxillofacial Surgery, Nihon University School of Dentistry at Matsudo
| | - Hiroyasu Tsukahara
- Department of Maxillofacial Surgery, Nihon University School of Dentistry at Matsudo
| | - Masaaki Suemitsu
- Research Institute of oral science, Nihon University School of Dentistry at Matsudo
- Department of oral pathology, Nihon University School of Dentistry at Matsudo
| | - Noboru Kuboyama
- Research Institute of oral science, Nihon University School of Dentistry at Matsudo
| | - Kayo Kuyama
- Research Institute of oral science, Nihon University School of Dentistry at Matsudo
- Department of oral pathology, Nihon University School of Dentistry at Matsudo
| | - Toshirou Kondoh
- Research Institute of oral science, Nihon University School of Dentistry at Matsudo
- Department of Maxillofacial Surgery, Nihon University School of Dentistry at Matsudo
| |
Collapse
|
22
|
Andl T, Le Bras GF, Richards NF, Allison GL, Loomans HA, Washington MK, Revetta F, Lee RK, Taylor C, Moses HL, Andl CD. Concerted loss of TGFβ-mediated proliferation control and E-cadherin disrupts epithelial homeostasis and causes oral squamous cell carcinoma. Carcinogenesis 2014; 35:2602-10. [PMID: 25233932 DOI: 10.1093/carcin/bgu194] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022] Open
Abstract
Although the etiology of squamous cell carcinomas of the oral mucosa is well understood, the cellular origin and the exact molecular mechanisms leading to their formation are not. Previously, we observed the coordinated loss of E-cadherin (CDH1) and transforming growth factor beta receptor II (TGFBR2) in esophageal squamous tumors. To investigate if the coordinated loss of Cdh1 and Tgfbr2 is sufficient to induce tumorigenesis in vivo, we developed two mouse models targeting ablation of both genes constitutively or inducibly in the oral-esophageal epithelium. We show that the loss of both Cdh1 and Tgfbr2 in both models is sufficient to induce squamous cell carcinomas with animals succumbing to the invasive disease by 18 months of age. Advanced tumors have the ability to invade regional lymph nodes and to establish distant pulmonary metastasis. The mouse tumors showed molecular characteristics of human tumors such as overexpression of Cyclin D1. We addressed the question whether TGFβ signaling may target known stem cell markers and thereby influence tumorigenesis. From our mouse and human models, we conclude that TGFβ signaling regulates key aspects of stemness and quiescence in vitro and in vivo. This provides a new explanation for the importance of TGFβ in mucosal homeostasis.
Collapse
Affiliation(s)
- Thomas Andl
- Division of Dermatology, Department of Medicine, Department of Surgery, Department of Cancer Biology, Vanderbilt Ingram Cancer Center, Vanderbilt Digestive Disease Center and Department of Pathology, Vanderbilt University Medical Center, Nashville, TN 37232-6840, USA
| | | | | | | | | | - M Kay Washington
- Vanderbilt Ingram Cancer Center, Vanderbilt Digestive Disease Center and Department of Pathology, Vanderbilt University Medical Center, Nashville, TN 37232-6840, USA
| | - Frank Revetta
- Vanderbilt Ingram Cancer Center, Vanderbilt Digestive Disease Center and Department of Pathology, Vanderbilt University Medical Center, Nashville, TN 37232-6840, USA
| | | | | | - Harold L Moses
- Department of Cancer Biology, Vanderbilt Ingram Cancer Center
| | - Claudia D Andl
- Department of Surgery, Department of Cancer Biology, Vanderbilt Ingram Cancer Center, Vanderbilt Digestive Disease Center and
| |
Collapse
|
23
|
Hall BE, Wankhade UD, Konkel JE, Cherukuri K, Nagineni CN, Flanders KC, Arany PR, Chen W, Rane SG, Kulkarni AB. Transforming growth factor-β3 (TGF-β3) knock-in ameliorates inflammation due to TGF-β1 deficiency while promoting glucose tolerance. J Biol Chem 2013; 288:32074-92. [PMID: 24056369 DOI: 10.1074/jbc.m113.480764] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
Three homologues of TGF-β exist in mammals as follows: TGF-β1, TGF-β2, and TGF-β3. All three proteins share high homology in their amino acid sequence, yet each TGF-β isoform has unique heterologous motifs that are highly conserved during evolution. Although these TGF-β proteins share similar properties in vitro, isoform-specific properties have been suggested through in vivo studies and by the unique phenotypes for each TGF-β knock-out mouse. To test our hypothesis that each of these homologues has nonredundant functions, and to identify such isoform-specific roles, we genetically exchanged the coding sequence of the mature TGF-β1 ligand with a sequence from TGF-β3 using targeted recombination to create chimeric TGF-β1/3 knock-in mice (TGF-β1(Lβ3/Lβ3)). In the TGF-β1(Lβ3/Lβ3) mouse, localization and activation still occur through the TGF-β1 latent associated peptide, but cell signaling is triggered through the TGF-β3 ligand that binds to TGF-β receptors. Unlike TGF-β1(-/-) mice, the TGF-β1(Lβ3/Lβ3) mice show neither embryonic lethality nor signs of multifocal inflammation, demonstrating that knock-in of the TGF-β3 ligand can prevent the vasculogenesis defects and autoimmunity associated with TGF-β1 deficiency. However, the TGF-β1(Lβ3/Lβ3) mice have a shortened life span and display tooth and bone defects, indicating that the TGF-β homologues are not completely interchangeable. Remarkably, the TGF-β1(Lβ3/Lβ3) mice display an improved metabolic phenotype with reduced body weight gain and enhanced glucose tolerance by induction of beneficial changes to the white adipose tissue compartment. These findings reveal both redundant and unique nonoverlapping functional diversity in TGF-β isoform signaling that has relevance to the design of therapeutics aimed at targeting the TGF-β pathway in human disease.
Collapse
Affiliation(s)
- Bradford E Hall
- From the Functional Genomics Section, Laboratory of Cell and Developmental Biology
| | | | | | | | | | | | | | | | | | | |
Collapse
|
24
|
Inhibiting TGF-β activity improves respiratory function in mdx mice. THE AMERICAN JOURNAL OF PATHOLOGY 2011; 178:2611-21. [PMID: 21641384 DOI: 10.1016/j.ajpath.2011.02.024] [Citation(s) in RCA: 66] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/19/2010] [Revised: 01/26/2011] [Accepted: 02/03/2011] [Indexed: 11/23/2022]
Abstract
Respiratory function is the main cause of mortality in patients with Duchenne muscular dystrophy (DMD). Elevated levels of TGF-β play a key role in the pathophysiology of DMD. To determine whether therapeutic attenuation of TGF-β signaling improves respiratory function, mdx mice were treated from 2 weeks of age to 2 months or 9 months of age with either 1D11 (a neutralizing antibody to all three isoforms of TGF-β), losartan (an angiotensin receptor antagonist), or a combination of the two agents. Respiratory function was measured in nonanesthetized mice by plethysmography. The 9-month-old mdx mice had elevated Penh values and decreased breathing frequency, due primarily to decreased inspiratory flow rate. All treatments normalized Penh values and increased peak inspiratory flow, leading to decreased inspiration times and breathing frequency. Additionally, forelimb grip strength was improved after 1D11 treatment at both 2 and 9 months of age, whereas, losartan improved grip strength only at 2 months. Decreased serum creatine kinase levels (significant improvement for all groups), increased diaphragm muscle fiber density, and decreased hydroxyproline levels (significant improvement for 1D11 only) also suggested improved muscle function after treatment. For all endpoints, 1D11 was equivalent or superior to losartan; coadministration of the two agents was not superior to 1D11 alone. In conclusion, TGF-β antagonism may be a useful therapeutic approach for treating DMD patients.
Collapse
|
25
|
Kim DY, Kwon EY, Hong GU, Lee YS, Lee SH, Ro JY. Cigarette smoke exacerbates mouse allergic asthma through Smad proteins expressed in mast cells. Respir Res 2011; 12:49. [PMID: 21496353 PMCID: PMC3098800 DOI: 10.1186/1465-9921-12-49] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2010] [Accepted: 04/18/2011] [Indexed: 11/10/2022] Open
Abstract
Background Many studies have found that smoking reduces lung function, but the relationship between cigarette smoke and allergic asthma has not been clearly elucidated, particularly the role of mast cells. This study aimed to investigate the effects of smoke exposure on allergic asthma and its association with mast cells. Methods BALB/c mice were sensitized and challenged by OVA to induce asthma, and bone marrow-derived mast cells (BMMCs) were stimulated with antigen/antibody reaction. Mice or BMMCs were exposed to cigarette smoke or CSE solution for 1 mo or 6 h, respectively. The recruitment of inflammatory cells into BAL fluid or lung tissues was determined by Diff-Quik or H&E staining, collagen deposition by Sircol assay, penh values by a whole-body plethysmography, co-localization of tryptase and Smad3 by immunohistochemistry, IgE and TGF-β level by ELISA, expressions of Smads proteins, activities of signaling molecules, or TGF-β mRNA by immunoblotting and RT-PCR. Results Cigarette smoke enhanced OVA-specific IgE levels, penh values, recruitment of inflammatory cells including mast cells, expressions of smad family, TGF-β mRNA and proteins, and cytokines, phosphorylations of Smad2 and 3, and MAP kinases, co-localization of tryptase and Smad3, and collagen deposition more than those of BAL cells and lung tissues of OVA-induced allergic mice. CSE solution pretreatment enhanced expressions of TGF-β, Smad3, activities of MAP kinases, NF-κB/AP-1 or PAI-1 more than those of activated-BMMCs. Conclusions The data suggest that smoke exposure enhances antigen-induced mast cell activation via TGF-β/Smad signaling pathways in mouse allergic asthma, and that it exacerbates airway inflammation and remodeling.
Collapse
Affiliation(s)
- Dae Yong Kim
- Department of Pharmacology and Samsung Biomedical Research Institute, Sungkyunkwan University School of Medicine, Suwon 440-726, Korea
| | | | | | | | | | | |
Collapse
|