1
|
Skingen VE, Salberg UB, Hompland T, Fjeldbo CS, Helgeland H, Frikstad KAM, Ragnum HB, Vlatkovic L, Hole KH, Seierstad T, Lyng H. Spatial analysis of microRNA regulation at defined tumor hypoxia levels reveals biological traits of aggressive prostate cancer. J Pathol 2024; 264:270-283. [PMID: 39329425 DOI: 10.1002/path.6344] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2024] [Revised: 06/27/2024] [Accepted: 07/29/2024] [Indexed: 09/28/2024]
Abstract
Mechanisms regulating the gene expression program at different hypoxia severity levels in patient tumors are not understood. We aimed to determine microRNA (miRNA) regulation of this program at defined hypoxia levels from moderate to severe in prostate cancer. Biopsies from 95 patients were used, where 83 patients received the hypoxia marker pimonidazole before prostatectomy. Forty hypoxia levels were extracted from pimonidazole-stained histological sections and correlated with miRNA and gene expression profiles determined by RNA sequencing and Illumina bead arrays. This identified miRNAs associated with moderate (n = 7) and severe (n = 28) hypoxia and predicted their target genes. The scores of miRNAs or target genes showed prognostic significance, as validated in an external cohort of 417 patients. The target genes showed enrichment of gene sets for cell proliferation and MYC activation at all hypoxia levels and PTEN inactivation at severe hypoxia. This was confirmed by RT-qPCR for MYC and PTEN, by Ki67 immunohistochemistry, and by gene set analysis in an external cohort. To assess whether miRNA regulation occurred within the predicted hypoxic regions, a method to quantify co-localization of multiple histopathology parameters at defined hypoxia levels was applied. A high Ki67 proliferation index co-localized significantly with hypoxia at all levels. The co-localization index was strongly associated with poor prognosis. Absence of PTEN staining co-localized significantly with severe hypoxia. The scores for miRNAs correlated with the co-localization index for Ki67 staining and hypoxia, consistent with miRNA regulation within the overlapping regions. This was confirmed by showing miR-210-3p expression within severe hypoxia by in situ hybridization. Cell line experiments (22Rv1, PC3) were conducted to determine whether miRNAs and target genes were regulated directly by hypoxia. Most of them were hypoxia-unresponsive, and probably regulated by other mechanisms such as MYC activation. In conclusion, in aggressive, hypoxic prostate tumors, cancer cells exhibit different proliferative gene expression programs that is regulated by miRNAs and depend on whether the cells reside in moderate or severe hypoxic regions. © 2024 The Author(s). The Journal of Pathology published by John Wiley & Sons Ltd on behalf of The Pathological Society of Great Britain and Ireland.
Collapse
Affiliation(s)
- Vilde E Skingen
- Department of Radiation Biology, Institute for Cancer Research, Oslo University Hospital, Oslo, Norway
- Department of Physics, University of Oslo, Oslo, Norway
| | - Unn Beate Salberg
- Department of Radiation Biology, Institute for Cancer Research, Oslo University Hospital, Oslo, Norway
- Institute of Clinical Medicine, University of Oslo, Oslo, Norway
| | - Tord Hompland
- Department of Radiation Biology, Institute for Cancer Research, Oslo University Hospital, Oslo, Norway
| | - Christina S Fjeldbo
- Department of Radiation Biology, Institute for Cancer Research, Oslo University Hospital, Oslo, Norway
| | - Hanna Helgeland
- Department of Radiation Biology, Institute for Cancer Research, Oslo University Hospital, Oslo, Norway
| | - Kari-Anne M Frikstad
- Department of Radiation Biology, Institute for Cancer Research, Oslo University Hospital, Oslo, Norway
| | - Harald B Ragnum
- Department of Hematology and Oncology, Telemark Hospital Trust, Skien, Norway
| | | | - Knut Håkon Hole
- Institute of Clinical Medicine, University of Oslo, Oslo, Norway
- Division of Radiology and Nuclear Medicine, Oslo University Hospital, Oslo, Norway
| | - Therese Seierstad
- Division of Radiology and Nuclear Medicine, Oslo University Hospital, Oslo, Norway
| | - Heidi Lyng
- Department of Radiation Biology, Institute for Cancer Research, Oslo University Hospital, Oslo, Norway
- Department of Physics, University of Oslo, Oslo, Norway
| |
Collapse
|
2
|
Besso MJ, Bitto V, Koi L, Wijaya Hadiwikarta W, Conde-Lopez C, Euler-Lange R, Bonrouhi M, Schneider K, Linge A, Krause M, Baumann M, Kurth I. Transcriptomic and epigenetic landscape of nimorazole-enhanced radiochemotherapy in head and neck cancer. Radiother Oncol 2024; 199:110348. [PMID: 38823583 DOI: 10.1016/j.radonc.2024.110348] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2023] [Revised: 04/27/2024] [Accepted: 05/20/2024] [Indexed: 06/03/2024]
Abstract
BACKGROUND Hypoxia remains a challenge for the therapeutic management of head and neck squamous cell carcinoma (HNSCC). The combination of radiotherapy with nimorazole has shown treatment benefit in HNSCC, but the precise underlying molecular mechanisms remain unclear. PURPOSE To assess and to characterize the transcriptomic/epigenetic landscape of HNSCC tumor models showing differential therapeutic response to fractionated radiochemotherapy (RCTx) combined with nimorazole. MATERIALS/METHODS Bulk RNA-sequencing and DNA methylation experiments were conducted using untreated and treated HNSCC xenografts after 10 fractions of RCTx with and without nimorazole. These tumor models (FaDu, SAS, Cal33, SAT and UT-SCC-45) previously showed a heterogeneous response to RCTx with nimorazole. The prognostic impact of candidate genes was assessed using clinical and gene expression data from HNSCC patients treated with primary RCTx within the DKTK-ROG. RESULTS Nimorazole responder and non-responder tumor models showed no differences in hypoxia gene signatures However, non-responder models showed upregulation of metabolic pathways. From that, a subset of 15 differentially expressed genes stratified HNSCC patients into low and high-risk groups with distinct outcome. CONCLUSION In the present study, we found that nimorazole non-responder models were characterized by upregulation of genes involved in Retinol metabolism and xenobiotic metabolic process pathways, which might contribute to identify mechanisms of resistance to nitroimidazole compounds and potentially expand the repertoire of therapeutic options to treat HNSCC.
Collapse
Affiliation(s)
- María José Besso
- German Cancer Research Center (DKFZ) Heidelberg, Division of Radiooncology Radiobiology, Germany.
| | - Verena Bitto
- German Cancer Research Center (DKFZ) Heidelberg, Division of Radiooncology Radiobiology, Germany; German Cancer Research Center (DKFZ) Heidelberg, Division of Applied Bioinformatics, Germany; Faculty for Mathematics and Computer Science, Heidelberg University, Germany; HIDSS4Health - Helmholtz Information and Data Science School for Health, Karlsruhe/Heidelberg, Germany
| | - Lydia Koi
- OncoRay - National Center for Radiation Research in Oncology, Faculty of Medicine and University Hospital Carl Gustav Carus, Technische Universität Dresden, Helmholtz-Zentrum Dresden - Rossendorf, Dresden, Germany; Department of Radiotherapy and Radiation Oncology, Faculty of Medicine and University Hospital Carl Gustav Carus, Technische Universität Dresden, Dresden, Germany; Helmholtz-Zentrum Dresden - Rossendorf, Institute of Radiooncology - OncoRay, Dresden, Germany
| | - Wahyu Wijaya Hadiwikarta
- German Cancer Research Center (DKFZ) Heidelberg, Division of Radiooncology Radiobiology, Germany; National Center for Radiation Research in Oncology (NCRO), Heidelberg Institute for Radiation Oncology (HIRO), Heidelberg, Germany
| | - Cristina Conde-Lopez
- German Cancer Research Center (DKFZ) Heidelberg, Division of Radiooncology Radiobiology, Germany
| | - Rosemarie Euler-Lange
- German Cancer Research Center (DKFZ) Heidelberg, Division of Radiooncology Radiobiology, Germany
| | - Mahnaz Bonrouhi
- German Cancer Research Center (DKFZ) Heidelberg, Division of Radiooncology Radiobiology, Germany
| | - Karolin Schneider
- German Cancer Research Center (DKFZ) Heidelberg, Division of Radiooncology Radiobiology, Germany
| | - Annett Linge
- OncoRay - National Center for Radiation Research in Oncology, Faculty of Medicine and University Hospital Carl Gustav Carus, Technische Universität Dresden, Helmholtz-Zentrum Dresden - Rossendorf, Dresden, Germany; Department of Radiotherapy and Radiation Oncology, Faculty of Medicine and University Hospital Carl Gustav Carus, Technische Universität Dresden, Dresden, Germany; German Cancer Research Center (DKFZ), and German Cancer Consortium (DKTK), Partner Site Dresden, Heidelberg, Germany; National Center for Tumor Diseases (NCT), Partner Site Dresden, Germany; Faculty of Medicine and University Hospital Carl Gustav Carus, Technische Universität Dresden, and Helmholtz Association / Helmholtz-Zentrum Dresden Rossendorf (HZDR), Dresden, Germany
| | - Mechthild Krause
- OncoRay - National Center for Radiation Research in Oncology, Faculty of Medicine and University Hospital Carl Gustav Carus, Technische Universität Dresden, Helmholtz-Zentrum Dresden - Rossendorf, Dresden, Germany; Department of Radiotherapy and Radiation Oncology, Faculty of Medicine and University Hospital Carl Gustav Carus, Technische Universität Dresden, Dresden, Germany; Helmholtz-Zentrum Dresden - Rossendorf, Institute of Radiooncology - OncoRay, Dresden, Germany; German Cancer Research Center (DKFZ), and German Cancer Consortium (DKTK), Partner Site Dresden, Heidelberg, Germany; National Center for Tumor Diseases (NCT), Partner Site Dresden, Germany; Faculty of Medicine and University Hospital Carl Gustav Carus, Technische Universität Dresden, and Helmholtz Association / Helmholtz-Zentrum Dresden Rossendorf (HZDR), Dresden, Germany
| | - Michael Baumann
- German Cancer Research Center (DKFZ) Heidelberg, Division of Radiooncology Radiobiology, Germany; OncoRay - National Center for Radiation Research in Oncology, Faculty of Medicine and University Hospital Carl Gustav Carus, Technische Universität Dresden, Helmholtz-Zentrum Dresden - Rossendorf, Dresden, Germany; German Cancer Research Center (DKFZ), and German Cancer Consortium (DKTK), Core Center Heidelberg, Heidelberg, Germany; National Center for Radiation Research in Oncology (NCRO), Heidelberg Institute for Radiation Oncology (HIRO), Heidelberg, Germany
| | - Ina Kurth
- German Cancer Research Center (DKFZ) Heidelberg, Division of Radiooncology Radiobiology, Germany; German Cancer Research Center (DKFZ), and German Cancer Consortium (DKTK), Core Center Heidelberg, Heidelberg, Germany; National Center for Radiation Research in Oncology (NCRO), Heidelberg Institute for Radiation Oncology (HIRO), Heidelberg, Germany
| |
Collapse
|
3
|
Ortmann BM, Taylor CT, Rocha S. Hypoxia research, where to now? Trends Biochem Sci 2024; 49:573-582. [PMID: 38599898 DOI: 10.1016/j.tibs.2024.03.008] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2023] [Revised: 03/01/2024] [Accepted: 03/18/2024] [Indexed: 04/12/2024]
Abstract
Investigating how cells and organisms sense and respond to O2 levels is essential to our understanding of physiology and pathology. This field has advanced considerably since the discovery of the major transcription factor family, hypoxia-inducible factor (HIF), and the enzymes that control its levels: prolyl hydroxylases (PHDs). However, with its expansion, new complexities have emerged. Herein we highlight three main areas where, in our opinion, the research community could direct some of their attention. These include non-transcriptional roles of HIFs, specificity and O2 sensitivity of 2-oxoglutarate-dependent dioxygenases (2-OGDDs), and new tools and methods to detect O2 concentrations in cells and organs. A greater understanding of these areas would answer big questions and help drive our knowledge of cellular responses to hypoxia forward.
Collapse
Affiliation(s)
- Brian M Ortmann
- Wolfson Childhood Cancer Research Centre, Newcastle University, Newcastle upon Tyne, UK.
| | - Cormac T Taylor
- School of Medicine and Medical Sciences, University College Dublin, Dublin, Ireland.
| | - Sonia Rocha
- Institute of Systems Molecular and Integrative Biology, University of Liverpool, Liverpool, UK.
| |
Collapse
|
4
|
Gupta VK, Vaishnavi VV, Arrieta-Ortiz ML, P S A, K M J, Jeyasankar S, Raghunathan V, Baliga NS, Agarwal R. 3D Hydrogel Culture System Recapitulates Key Tuberculosis Phenotypes and Demonstrates Pyrazinamide Efficacy. Adv Healthc Mater 2024:e2304299. [PMID: 38655817 PMCID: PMC7616495 DOI: 10.1002/adhm.202304299] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2023] [Revised: 03/29/2024] [Indexed: 04/26/2024]
Abstract
The mortality caused by tuberculosis (TB) infections is a global concern, and there is a need to improve understanding of the disease. Current in vitro infection models to study the disease have limitations such as short investigation durations and divergent transcriptional signatures. This study aims to overcome these limitations by developing a 3D collagen culture system that mimics the biomechanical and extracellular matrix (ECM) of lung microenvironment (collagen fibers, stiffness comparable to in vivo conditions) as the infection primarily manifests in the lungs. The system incorporates Mycobacterium tuberculosis (Mtb) infected human THP-1 or primary monocytes/macrophages. Dual RNA sequencing reveals higher mammalian gene expression similarity with patient samples than 2D macrophage infections. Similarly, bacterial gene expression more accurately recapitulates in vivo gene expression patterns compared to bacteria in 2D infection models. Key phenotypes observed in humans, such as foamy macrophages and mycobacterial cords, are reproduced in the model. This biomaterial system overcomes challenges associated with traditional platforms by modulating immune cells and closely mimicking in vivo infection conditions, including showing efficacy with clinically relevant concentrations of anti-TB drug pyrazinamide, not seen in any other in vitro infection model, making it reliable and readily adoptable for tuberculosis studies and drug screening.
Collapse
Affiliation(s)
- Vishal K Gupta
- Department of Bioengineering, Indian Institute of Science, CV Raman Road, Bengaluru, Karnataka, 560012, India
| | - Vijaya V Vaishnavi
- Department of Bioengineering, Indian Institute of Science, CV Raman Road, Bengaluru, Karnataka, 560012, India
| | | | - Abhirami P S
- Department of Bioengineering, Indian Institute of Science, CV Raman Road, Bengaluru, Karnataka, 560012, India
| | - Jyothsna K M
- Department of Electrical Communication Engineering, Indian Institute of Science, CV Raman Road, Bengaluru, Karnataka, 560012, India
| | - Sharumathi Jeyasankar
- Department of Bioengineering, Indian Institute of Science, CV Raman Road, Bengaluru, Karnataka, 560012, India
| | - Varun Raghunathan
- Department of Electrical Communication Engineering, Indian Institute of Science, CV Raman Road, Bengaluru, Karnataka, 560012, India
| | - Nitin S Baliga
- Institute of Systems Biology, 401 Terry Ave N, Seattle, WA, 98109, USA
| | - Rachit Agarwal
- Department of Bioengineering, Indian Institute of Science, CV Raman Road, Bengaluru, Karnataka, 560012, India
| |
Collapse
|
5
|
Sahovaler A, Valic MS, Townson JL, Chan HH, Zheng M, Tzelnick S, Mondello T, Pener-Tessler A, Eu D, El-Sayes A, Ding L, Chen J, Douglas CM, Weersink R, Muhanna N, Zheng G, Irish JC. Nanoparticle-mediated Photodynamic Therapy as a Method to Ablate Oral Cavity Squamous Cell Carcinoma in Preclinical Models. CANCER RESEARCH COMMUNICATIONS 2024; 4:796-810. [PMID: 38421899 PMCID: PMC10941731 DOI: 10.1158/2767-9764.crc-23-0269] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/16/2023] [Revised: 12/05/2023] [Accepted: 02/27/2024] [Indexed: 03/02/2024]
Abstract
Photodynamic therapy (PDT) is a tissue ablation technique able to selectively target tumor cells by activating the cytotoxicity of photosensitizer dyes with light. PDT is nonsurgical and tissue sparing, two advantages for treatments in anatomically complex disease sites such as the oral cavity. We have previously developed PORPHYSOME (PS) nanoparticles assembled from chlorin photosensitizer-containing building blocks (∼94,000 photosensitizers per particle) and capable of potent PDT. In this study, we demonstrate the selective uptake and curative tumor ablation of PS-enabled PDT in three preclinical models of oral cavity squamous cell carcinoma (OCSCC): biologically relevant subcutaneous Cal-33 (cell line) and MOC22 (syngeneic) mouse models, and an anatomically relevant orthotopic VX-2 rabbit model. Tumors selectively uptake PS (10 mg/kg, i.v.) with 6-to 40-fold greater concentration versus muscle 24 hours post-injection. Single PS nanoparticle-mediated PDT (PS-PDT) treatment (100 J/cm2, 100 mW/cm2) of Cal-33 tumors yielded significant apoptosis in 65.7% of tumor cells. Survival studies following PS-PDT treatments demonstrated 90% (36/40) overall response rate across all three tumor models. Complete tumor response was achieved in 65% of Cal-33 and 91% of MOC22 tumor mouse models 14 days after PS-PDT, and partial responses obtained in 25% and 9% of Cal-33 and MOC22 tumors, respectively. In buccal VX-2 rabbit tumors, combined surface and interstitial PS-PDT (200 J total) yielded complete responses in only 60% of rabbits 6 weeks after a single treatment whereas three repeated weekly treatments with PS-PDT (200 J/week) achieved complete ablation in 100% of tumors. PS-PDT treatments were well tolerated by animals with no treatment-associated toxicities and excellent cosmetic outcomes. SIGNIFICANCE PS-PDT is a safe and repeatable treatment modality for OCSCC ablation. PS demonstrated tumor selective uptake and PS-PDT treatments achieved reproducible efficacy and effectiveness in multiple tumor models superior to other clinically tested photosensitizer drugs. Cosmetic and functional outcomes were excellent, and no clinically significant treatment-associated toxicities were detected. These results are enabling of window of opportunity trials for fluorescence-guided PS-PDT in patients with early-stage OCSCC scheduled for surgery.
Collapse
Affiliation(s)
- Axel Sahovaler
- Department of Otolaryngology–Head and Neck Surgery, University of Toronto, Toronto, Ontario, Canada
- TECHNA Institute, Guided Therapeutics (GTx) Program, University Health Network, Toronto, Ontario, Canada
- Princess Margaret Cancer Centre, University Health Network, Toronto, Ontario, Canada
| | - Michael S. Valic
- Princess Margaret Cancer Centre, University Health Network, Toronto, Ontario, Canada
- Institute of Biomedical Engineering (BME), University of Toronto, Toronto, Ontario, Canada
| | - Jason L. Townson
- TECHNA Institute, Guided Therapeutics (GTx) Program, University Health Network, Toronto, Ontario, Canada
- Princess Margaret Cancer Centre, University Health Network, Toronto, Ontario, Canada
| | - Harley H.L. Chan
- TECHNA Institute, Guided Therapeutics (GTx) Program, University Health Network, Toronto, Ontario, Canada
- Princess Margaret Cancer Centre, University Health Network, Toronto, Ontario, Canada
| | - Mark Zheng
- Princess Margaret Cancer Centre, University Health Network, Toronto, Ontario, Canada
| | - Sharon Tzelnick
- Department of Otolaryngology–Head and Neck Surgery, University of Toronto, Toronto, Ontario, Canada
- TECHNA Institute, Guided Therapeutics (GTx) Program, University Health Network, Toronto, Ontario, Canada
- Princess Margaret Cancer Centre, University Health Network, Toronto, Ontario, Canada
| | - Tiziana Mondello
- Department of Otolaryngology–Head and Neck Surgery, University of Toronto, Toronto, Ontario, Canada
- TECHNA Institute, Guided Therapeutics (GTx) Program, University Health Network, Toronto, Ontario, Canada
- Princess Margaret Cancer Centre, University Health Network, Toronto, Ontario, Canada
| | - Alon Pener-Tessler
- Department of Otolaryngology–Head and Neck Surgery, University of Toronto, Toronto, Ontario, Canada
- TECHNA Institute, Guided Therapeutics (GTx) Program, University Health Network, Toronto, Ontario, Canada
- Princess Margaret Cancer Centre, University Health Network, Toronto, Ontario, Canada
| | - Donovan Eu
- Department of Otolaryngology–Head and Neck Surgery, University of Toronto, Toronto, Ontario, Canada
- TECHNA Institute, Guided Therapeutics (GTx) Program, University Health Network, Toronto, Ontario, Canada
- Princess Margaret Cancer Centre, University Health Network, Toronto, Ontario, Canada
| | - Abdullah El-Sayes
- Princess Margaret Cancer Centre, University Health Network, Toronto, Ontario, Canada
| | - Lili Ding
- Princess Margaret Cancer Centre, University Health Network, Toronto, Ontario, Canada
| | - Juan Chen
- Princess Margaret Cancer Centre, University Health Network, Toronto, Ontario, Canada
| | - Catriona M. Douglas
- Department of Otolaryngology–Head and Neck Surgery, University of Toronto, Toronto, Ontario, Canada
- TECHNA Institute, Guided Therapeutics (GTx) Program, University Health Network, Toronto, Ontario, Canada
- Department of Otolaryngology–Head and Neck Surgery, Queen Elizabeth University Hospital, Glasgow, United Kingdom
| | - Robert Weersink
- TECHNA Institute, Guided Therapeutics (GTx) Program, University Health Network, Toronto, Ontario, Canada
- Princess Margaret Cancer Centre, University Health Network, Toronto, Ontario, Canada
- Department of Medical Biophysics, University of Toronto, Toronto, Ontario, Canada
| | - Nidal Muhanna
- Department of Otolaryngology–Head and Neck Surgery, University of Toronto, Toronto, Ontario, Canada
- TECHNA Institute, Guided Therapeutics (GTx) Program, University Health Network, Toronto, Ontario, Canada
- Department of Otolaryngology–Head and Neck Surgery, Tel Aviv Sourasky Medical Centre, Tel Aviv University, Tel Aviv, Israel
| | - Gang Zheng
- Princess Margaret Cancer Centre, University Health Network, Toronto, Ontario, Canada
- Institute of Biomedical Engineering (BME), University of Toronto, Toronto, Ontario, Canada
- Department of Medical Biophysics, University of Toronto, Toronto, Ontario, Canada
| | - Jonathan C. Irish
- Department of Otolaryngology–Head and Neck Surgery, University of Toronto, Toronto, Ontario, Canada
- TECHNA Institute, Guided Therapeutics (GTx) Program, University Health Network, Toronto, Ontario, Canada
- Princess Margaret Cancer Centre, University Health Network, Toronto, Ontario, Canada
| |
Collapse
|
6
|
Bigos KJA, Quiles CG, Lunj S, Smith DJ, Krause M, Troost EGC, West CM, Hoskin P, Choudhury A. Tumour response to hypoxia: understanding the hypoxic tumour microenvironment to improve treatment outcome in solid tumours. Front Oncol 2024; 14:1331355. [PMID: 38352889 PMCID: PMC10861654 DOI: 10.3389/fonc.2024.1331355] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2023] [Accepted: 01/08/2024] [Indexed: 02/16/2024] Open
Abstract
Hypoxia is a common feature of solid tumours affecting their biology and response to therapy. One of the main transcription factors activated by hypoxia is hypoxia-inducible factor (HIF), which regulates the expression of genes involved in various aspects of tumourigenesis including proliferative capacity, angiogenesis, immune evasion, metabolic reprogramming, extracellular matrix (ECM) remodelling, and cell migration. This can negatively impact patient outcomes by inducing therapeutic resistance. The importance of hypoxia is clearly demonstrated by continued research into finding clinically relevant hypoxia biomarkers, and hypoxia-targeting therapies. One of the problems is the lack of clinically applicable methods of hypoxia detection, and lack of standardisation. Additionally, a lot of the methods of detecting hypoxia do not take into consideration the complexity of the hypoxic tumour microenvironment (TME). Therefore, this needs further elucidation as approximately 50% of solid tumours are hypoxic. The ECM is important component of the hypoxic TME, and is developed by both cancer associated fibroblasts (CAFs) and tumour cells. However, it is important to distinguish the different roles to develop both biomarkers and novel compounds. Fibronectin (FN), collagen (COL) and hyaluronic acid (HA) are important components of the ECM that create ECM fibres. These fibres are crosslinked by specific enzymes including lysyl oxidase (LOX) which regulates the stiffness of tumours and induces fibrosis. This is partially regulated by HIFs. The review highlights the importance of understanding the role of matrix stiffness in different solid tumours as current data shows contradictory results on the impact on therapeutic resistance. The review also indicates that further research is needed into identifying different CAF subtypes and their exact roles; with some showing pro-tumorigenic capacity and others having anti-tumorigenic roles. This has made it difficult to fully elucidate the role of CAFs within the TME. However, it is clear that this is an important area of research that requires unravelling as current strategies to target CAFs have resulted in worsened prognosis. The role of immune cells within the tumour microenvironment is also discussed as hypoxia has been associated with modulating immune cells to create an anti-tumorigenic environment. Which has led to the development of immunotherapies including PD-L1. These hypoxia-induced changes can confer resistance to conventional therapies, such as chemotherapy, radiotherapy, and immunotherapy. This review summarizes the current knowledge on the impact of hypoxia on the TME and its implications for therapy resistance. It also discusses the potential of hypoxia biomarkers as prognostic and predictive indictors of treatment response, as well as the challenges and opportunities of targeting hypoxia in clinical trials.
Collapse
Affiliation(s)
- Kamilla JA. Bigos
- Division of Cancer Sciences, University of Manchester, Manchester, United Kingdom
| | - Conrado G. Quiles
- Division of Cancer Sciences, University of Manchester, Manchester, United Kingdom
| | - Sapna Lunj
- Division of Cancer Sciences, University of Manchester, Manchester, United Kingdom
| | - Danielle J. Smith
- Division of Cancer Sciences, University of Manchester, Manchester, United Kingdom
| | - Mechthild Krause
- German Cancer Consortium (DKTK), partner site Dresden and German Cancer Research Center (DKFZ), Heidelberg, Germany
- Department of Radiation Oncology, Faculty of Medicine and University Hospital Carl Gustav Carus, Technische Universität Dresden, Dresden, Germany
- OncoRay – National Center for Radiation Research in Oncology, Faculty of Medicine and University Hospital Carl Gustav Carus, Technische Universität Dresden, Helmholtz-Zentrum Dresden - Rossendorf, Dresden, Germany
- Translational Radiooncology and Clinical Radiotherapy, Helmholtz-Zentrum Dresden - Rossendorf, Dresden, Germany
- Translational Radiation Oncology, National Center for Tumor Diseases (NCT), Partner Site Dresden, Dresden, Germany
- Translational Radiooncology and Clinical Radiotherapy and Image-guided High Precision Radiotherapy, German Cancer Research Center (DKFZ), Heidelberg, Germany
- Faculty of Medicine and University Hospital Carl Gustav Carus, Technische Universität Dresden, Dresden, Germany
- Translational Radiooncology and Clinical Radiotherapy and Image-guided High Precision Radiotherapy, Helmholtz Association / Helmholtz-Zentrum Dresden - Rossendorf (HZDR), Dresden, Germany
- School of Medicine, Technische Universitat Dresden, Dresden, Germany
| | - Esther GC. Troost
- OncoRay – National Center for Radiation Research in Oncology, Faculty of Medicine and University Hospital Carl Gustav Carus, Technische Universität Dresden, Helmholtz-Zentrum Dresden - Rossendorf, Dresden, Germany
- Translational Radiation Oncology, National Center for Tumor Diseases (NCT), Partner Site Dresden, Dresden, Germany
- Translational Radiooncology and Clinical Radiotherapy and Image-guided High Precision Radiotherapy, German Cancer Research Center (DKFZ), Heidelberg, Germany
- Faculty of Medicine and University Hospital Carl Gustav Carus, Technische Universität Dresden, Dresden, Germany
- Translational Radiooncology and Clinical Radiotherapy and Image-guided High Precision Radiotherapy, Helmholtz Association / Helmholtz-Zentrum Dresden - Rossendorf (HZDR), Dresden, Germany
- School of Medicine, Technische Universitat Dresden, Dresden, Germany
- Department of Radiotherapy and Radiation Oncology, Faculty of Medicine and University Hospital Carl Gustav Carus, Technische Universität Dresden, Dresden, Germany
- Institute of Radiooncology – OncoRay, Helmholtz-Zentrum Dresden-Rossendorf, Rossendorf, Germany
| | - Catharine M. West
- Division of Cancer Sciences, University of Manchester, Manchester Academic Health Science Centre, Christie Hospital, Manchester, United Kingdom
| | - Peter Hoskin
- Division of Cancer Sciences, University of Manchester, Manchester, United Kingdom
- Mount Vernon Cancer Centre, Northwood, United Kingdom
| | - Ananya Choudhury
- Division of Cancer Sciences, University of Manchester, Manchester, United Kingdom
- Christie Hospital NHS Foundation Trust, Manchester, Germany
| |
Collapse
|
7
|
Srivastava I, Moitra P, Brent KM, Wang K, Pandit S, Altun E, Pan D. Biodegradable and switchable near-infrared fluorescent probes for hypoxia detection. Nanomedicine (Lond) 2023; 18:1061-1073. [PMID: 37610080 DOI: 10.2217/nnm-2023-0095] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 08/24/2023] Open
Abstract
Aims: Among solid tumors, hypoxia is a common characteristic and responsible for chemotherapeutic resistance. Hypoxia-sensitive imaging probes are therefore essential for early tumor detection, growth monitoring and drug-response evaluation. Despite significant efforts, detecting hypoxic oxygen levels remains challenging. Materials & methods: This paper demonstrates the use of an amine-rich carbon dot probe functionalized with an imidazole group that exhibits reversible fluorescence switching in normoxic and hypoxic environments. Results & conclusion: We demonstrate the ability to emit near-infrared light only under hypoxic conditions. The probes are found to be biodegradable in the presence of human digestive enzymes such as lipase. Ex vivo tissue imaging experiments revealed promising near-infrared signals even at a depth of 5 mm for the probe under ex vivo imaging conditions.
Collapse
Affiliation(s)
- Indrajit Srivastava
- Department of Bioengineering, University of Illinois at Urbana-Champaign, Urbana, IL 61801, USA
- Mills Breast Cancer Institute, Carle Foundation Hospital, Urbana, IL 61801, USA
| | - Parikshit Moitra
- Department of Nuclear Engineering, The Pennsylvania State University, University Park, PA 16802, USA
| | - Kurtis M Brent
- Department of Bioengineering, University of Illinois at Urbana-Champaign, Urbana, IL 61801, USA
- Mills Breast Cancer Institute, Carle Foundation Hospital, Urbana, IL 61801, USA
| | - Kevin Wang
- Department of Bioengineering, University of Illinois at Urbana-Champaign, Urbana, IL 61801, USA
- Mills Breast Cancer Institute, Carle Foundation Hospital, Urbana, IL 61801, USA
| | - Subhendu Pandit
- Department of Bioengineering, University of Illinois at Urbana-Champaign, Urbana, IL 61801, USA
- Mills Breast Cancer Institute, Carle Foundation Hospital, Urbana, IL 61801, USA
| | - Esra Altun
- Department of Bioengineering, University of Illinois at Urbana-Champaign, Urbana, IL 61801, USA
- Mills Breast Cancer Institute, Carle Foundation Hospital, Urbana, IL 61801, USA
| | - Dipanjan Pan
- Department of Bioengineering, University of Illinois at Urbana-Champaign, Urbana, IL 61801, USA
- Mills Breast Cancer Institute, Carle Foundation Hospital, Urbana, IL 61801, USA
- Department of Nuclear Engineering, The Pennsylvania State University, University Park, PA 16802, USA
- Department of Materials Science and Engineering, The Pennsylvania State University, University Park, PA 16802, USA
- Huck Institutes of the Life Sciences, University Park, PA 16802, USA
| |
Collapse
|
8
|
Aboouf MA, Armbruster J, Guscetti F, Thiersch M, Boss A, Gödecke A, Winning S, Padberg C, Fandrey J, Kristiansen G, Bicker A, Hankeln T, Gassmann M, Gorr TA. Endogenous myoglobin expression in mouse models of mammary carcinoma reduces hypoxia and metastasis in PyMT mice. Sci Rep 2023; 13:7530. [PMID: 37161046 PMCID: PMC10170105 DOI: 10.1038/s41598-023-34614-w] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2022] [Accepted: 05/04/2023] [Indexed: 05/11/2023] Open
Abstract
Myoglobin (MB) is expressed in different cancer types and may act as a tumor suppressor in breast cancer. The mechanisms by which basal MB expression level impacts murine mammary tumorigenesis are unclear. We investigated how MB expression in breast cancer influences proliferation, metastasis, tumor hypoxia, and chemotherapy treatment in vivo. We crossed PyMT and WapCreTrp53flox mammary cancer mouse models that differed in tumor grade/type and onset of mammary carcinoma with MB knockout mice. The loss of MB in WapCre;Trp53flox mice did not affect tumor development and progression. On the other hand, loss of MB decreased tumor growth and increased tissue hypoxia as well as the number of lung metastases in PyMT mice. Furthermore, Doxorubicin therapy prevented the stronger metastatic propensity of MB-deficient tumors in PyMT mice. This suggests that, although MB expression predicts improved prognosis in breast cancer patients, MB-deficient tumors may still respond well to first-line therapies. We propose that determining the expression level of MB in malignant breast cancer biopsies will improve tumor stratification, outcome prediction, and personalized therapy in cancer patients.
Collapse
Affiliation(s)
- Mostafa A Aboouf
- Institute of Veterinary Physiology, Vetsuisse Faculty, University of Zurich, Winterthurerstrasse 260, 8057, Zurich, Switzerland
- Center for Clinical Studies, Vetsuisse Faculty, University of Zurich, 8057, Zurich, Switzerland
- Department of Biochemistry, Faculty of Pharmacy, Ain Shams University, Cairo, 11566, Egypt
- Zurich Center for Integrative Human Physiology (ZIHP), University of Zurich, 8057, Zurich, Switzerland
| | - Julia Armbruster
- Institute of Veterinary Physiology, Vetsuisse Faculty, University of Zurich, Winterthurerstrasse 260, 8057, Zurich, Switzerland
- Center for Clinical Studies, Vetsuisse Faculty, University of Zurich, 8057, Zurich, Switzerland
- Department of Biochemistry, Faculty of Pharmacy, Ain Shams University, Cairo, 11566, Egypt
| | - Franco Guscetti
- Institute of Veterinary Pathology, Vetsuisse Faculty, University of Zurich, 8057, Zurich, Switzerland
| | - Markus Thiersch
- Institute of Veterinary Physiology, Vetsuisse Faculty, University of Zurich, Winterthurerstrasse 260, 8057, Zurich, Switzerland
- Zurich Center for Integrative Human Physiology (ZIHP), University of Zurich, 8057, Zurich, Switzerland
| | - Andreas Boss
- Department of Diagnostic and Interventional Radiology, University Hospital Zurich, 8091, Zurich, Switzerland
| | - Axel Gödecke
- Institute of Cardiovascular Pathology, Medical Faculty, Heinrich Heine University, 40225, Düsseldorf, Germany
| | - Sandra Winning
- Institute for Physiology, University Duisburg-Essen, 47057, Essen, Germany
| | - Claudia Padberg
- Institute for Physiology, University Duisburg-Essen, 47057, Essen, Germany
| | - Joachim Fandrey
- Institute for Physiology, University Duisburg-Essen, 47057, Essen, Germany
| | - Glen Kristiansen
- Institute of Pathology, University Hospital Bonn, University of Bonn, 53127, Bonn, Germany
| | - Anne Bicker
- Institute of Organismic and Molecular Evolution, Molecular and Genome Analysis, Johannes Gutenberg University, 55099, Mainz, Germany
- University Medical Center Mainz, I. Medical Clinic, Langenbeckstr. 1, 55131, Mainz, Germany
| | - Thomas Hankeln
- Institute of Organismic and Molecular Evolution, Molecular and Genome Analysis, Johannes Gutenberg University, 55099, Mainz, Germany
| | - Max Gassmann
- Institute of Veterinary Physiology, Vetsuisse Faculty, University of Zurich, Winterthurerstrasse 260, 8057, Zurich, Switzerland
- Zurich Center for Integrative Human Physiology (ZIHP), University of Zurich, 8057, Zurich, Switzerland
| | - Thomas A Gorr
- Institute of Veterinary Physiology, Vetsuisse Faculty, University of Zurich, Winterthurerstrasse 260, 8057, Zurich, Switzerland.
| |
Collapse
|
9
|
Elming PB, Busk M, Wittenborn TR, Bussink J, Horsman MR, Lønbro S. The effect of single bout and prolonged aerobic exercise on tumor hypoxia in mice. J Appl Physiol (1985) 2023; 134:692-702. [PMID: 36727633 DOI: 10.1152/japplphysiol.00561.2022] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2022] [Revised: 01/17/2023] [Accepted: 01/30/2023] [Indexed: 02/03/2023] Open
Abstract
The objectives of this study were to investigate 1) the effect of acute aerobic exercise on tumor hypoxia and blood perfusion, 2) the impact of exercise intensity, 3) the duration of the effect, and 4) the effect of prolonged training on tumor hypoxia and tumor growth. Female CDF1 mice were inoculated with the C3H mammary carcinoma either in the mammary fat pad or subcutaneously in the back. For experiments on the effect of different intensities in a single exercise bout, mice were randomized to 30-min treadmill running at low-, moderate-, or high-intensity speeds or no exercise. To investigate the prolonged effect on hypoxia and tumor growth, tumor-bearing mice were randomized to no exercise (CON) or daily 30-min high-intensity exercise averaging 2 wk (EX). Tumor hypoxic fraction was quantified using the hypoxia marker Pimonidazole. Initially, high-intensity exercise reduced tumor hypoxic fraction by 37% compared with CON [P = 0.046; 95% confidence interval (CI): 0.1; 10.3] in fat pad tumors. Low- and moderate-intensity exercises did not. Following experiments investigating the duration of the effect-as well as experiments in mice with back tumors-failed to show any exercise-induced changes in hypoxia. Interestingly, prolonged daily training significantly reduced hypoxic fraction by 60% (P = 0.002; 95% CI: 2.5; 10.1) compared with CON. Despite diverging findings on the acute effect of exercise on hypoxia, our data indicate that if exercise has a diminishing effect, high-intensity exercise is needed. Prolonged training reduced tumor hypoxic fraction-cautiously suggesting a potential clinical potential.NEW & NOTEWORTHY This study provides novel information on the effects of acute and chronic exercise on tumor hypoxia in mice. In contrast to the few related existing studies, diverging findings on tumor hypoxia after acute exercise were observed, suggesting that tumor model and location should be considered in future studies. Highly significant reductions in tumor hypoxia following chronic high-intensity exercise propose a future clinical potential but this should be investigated in patients.
Collapse
Affiliation(s)
| | - Morten Busk
- Experimental Clinical Oncology, Department of Oncology, Aarhus University Hospital, Aarhus, Denmark
- Danish Centre for Particle Therapy, Aarhus University Hospital, Aarhus, Denmark
| | - Thomas Rea Wittenborn
- Experimental Clinical Oncology, Department of Oncology, Aarhus University Hospital, Aarhus, Denmark
| | - Johan Bussink
- Department of Radiation Oncology, Radboud University, Nijmegen Medical Centre, Nijmegen, The Netherlands
| | - Michael R Horsman
- Experimental Clinical Oncology, Department of Oncology, Aarhus University Hospital, Aarhus, Denmark
| | - Simon Lønbro
- Experimental Clinical Oncology, Department of Oncology, Aarhus University Hospital, Aarhus, Denmark
- Section for Sports Science, Department of Public Health, Aarhus University Hospital, Aarhus, Denmark
| |
Collapse
|
10
|
How the histological structure of some lung cancers shaped almost 70 years of radiobiology. Br J Cancer 2023; 128:407-412. [PMID: 36344595 PMCID: PMC9938174 DOI: 10.1038/s41416-022-02041-9] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2022] [Revised: 10/14/2022] [Accepted: 10/19/2022] [Indexed: 11/09/2022] Open
Abstract
Pivotal research led by Louis Harold Gray in the 1950s suggested that oxygen plays a vital role during radiotherapy. By proving that tumours have large necrotic cores due to hypoxia and that hypoxic cells require significantly larger doses of ionising radiation to achieve the same cell kill, Thomlinson and Gray inspired the subsequent decades of research into better defining the mechanistic role of molecular oxygen at the time of radiation. Ultimately, the work pioneered by Thomlinson and Gray led to numerous elegant studies which demonstrated that tumour hypoxia predicts for poor patient outcomes. Furthermore, this subsequently resulted in investigations into markers and measurement of hypoxia, as well as modification strategies. However, despite an abundance of pre-clinical data supporting hypoxia-targeted treatments, there is limited widespread application of hypoxia-targeted therapies routinely used in clinical practice. Significant contributing factors underpinning disappointing clinical trial results include the use of model systems which are more hypoxic than human tumours and a failure to stratify patients based on levels of hypoxia. However, translating the original findings of Thomlinson and Gray remains a research priority with the potential to significantly improve patient outcomes and specifically those receiving radiotherapy.
Collapse
|
11
|
Bourigault P, Skwarski M, Macpherson RE, Higgins GS, McGowan DR. Timing of hypoxia PET/CT imaging after 18F-fluoromisonidazole injection in non-small cell lung cancer patients. Sci Rep 2022; 12:21746. [PMID: 36526815 PMCID: PMC9758119 DOI: 10.1038/s41598-022-26199-7] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2022] [Accepted: 12/12/2022] [Indexed: 12/23/2022] Open
Abstract
Positron emission tomography (PET)/computed tomography (CT) using the radiotracer 18F-Fluoromisonidazole (FMISO) has been widely employed to image tumour hypoxia and is of interest to help develop novel hypoxia modifiers and guide radiation treatment planning. Yet, the optimal post-injection (p.i.) timing of hypoxic imaging remains questionable. Therefore, we investigated the correlation between hypoxia-related quantitative values in FMISO-PET acquired at 2 and 4 h p.i. in patients with non-small cell lung cancer (NSCLC). Patients with resectable NSCLC participated in the ATOM clinical trial (NCT02628080) which investigated the hypoxia modifying effects of atovaquone. Two-hour and four-hour FMISO PET/CT images acquired at baseline and pre-surgery visits (n = 58) were compared. Cohort 1 (n = 14) received atovaquone treatment, while cohort 2 (n = 15) did not. Spearman's rank correlation coefficients (ρ) assessed the relationship between hypoxia-related metrics, including standardised uptake value (SUV), tumour-to-blood ratio (TBR), and tumour hypoxic volume (HV) defined by voxels with TBR ≥ 1.4. As the primary imaging-related trial endpoint used to evaluate the action of atovaquone on tumour hypoxia in patients with NSCLC was change in tumour HV from baseline, this was also assessed in patients (n = 20) with sufficient baseline 2- and 4-h scan HV to reliably measure change (predefined as ≥ 1.5 mL). Tumours were divided into four subregions or distance categories: edge, outer, inner, and centre, using MATLAB. In tumours overall, strong correlation (P < 0.001) was observed for SUVmax ρ = 0.87, SUVmean ρ = 0.91, TBRmax ρ = 0.83 and TBRmean ρ = 0.81 between 2- and 4-h scans. Tumour HV was moderately correlated (P < 0.001) with ρ = 0.69 between 2- and 4-h scans. Yet, in tumour subregions, the correlation of HV decreased from the centre ρ = 0.71 to the edge ρ = 0.45 (P < 0.001). SUV, TBR, and HV values were consistently higher on 4-h scans than on 2-h scans, indicating better tracer-to-background contrast. For instance, for TBRmax, the mean, median, and interquartile range were 1.9, 1.7, and 1.6-2.0 2-h p.i., and 2.6, 2.4, and 2.0-3.0 4-h p.i., respectively. Our results support that FMISO-PET scans should be performed at 4 h p.i. to evaluate tumour hypoxia in NSCLC.Trial registration: ClinicalTrials.gov, NCT02628080. Registered 11/12/2015, https://clinicaltrials.gov/ct2/show/NCT02628080 .
Collapse
Affiliation(s)
| | - Michael Skwarski
- Department of Oncology, University of Oxford, Oxford, OX3 7DQ, UK
- Department of Oncology, Oxford University Hospitals NHS Foundation Trust, Oxford, UK
- Department of Clinical Oncology, Guy's and St Thomas' NHS Foundation Trust, London, UK
| | - Ruth E Macpherson
- Department of Radiology, Oxford University Hospitals NHS Foundation Trust, Oxford, UK
| | - Geoff S Higgins
- Department of Oncology, University of Oxford, Oxford, OX3 7DQ, UK
- Department of Oncology, Oxford University Hospitals NHS Foundation Trust, Oxford, UK
| | - Daniel R McGowan
- Department of Oncology, University of Oxford, Oxford, OX3 7DQ, UK.
- Department of Medical Physics and Clinical Engineering, Oxford University Hospitals NHS Foundation Trust, Oxford, UK.
| |
Collapse
|
12
|
Uehara-Watanabe N, Okuno-Ozeki N, Nakamura I, Nakata T, Nakai K, Yagi-Tomita A, Ida T, Yamashita N, Kamezaki M, Kirita Y, Matoba S, Tamagaki K, Kusaba T. Proximal tubular epithelia-specific transcriptomics of diabetic mice treated with dapagliflozin. Heliyon 2022; 8:e10615. [PMID: 36148274 PMCID: PMC9485043 DOI: 10.1016/j.heliyon.2022.e10615] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2021] [Revised: 02/26/2022] [Accepted: 09/07/2022] [Indexed: 11/26/2022] Open
Abstract
Based on recent clinical trials using sodium-glucose co-transporter 2 inhibitor (SGLT2i) demonstrating the significant improvement of outcomes of diabetic kidney disease (DKD), the paradigm shift from “glomerulocentric” to “tubule centric” pathophysiology in DKD progression has been highlighted. Several responsible mechanisms for renoprotective effects by SGLT2i have been proposed recently, but the changes in proximal tubule-specific gene expression by SGLT2i in diabetic mice have not been elucidated. We report the analysis of the proximal tubular-specific pathway, demonstrating the downregulation of oxidative phosphorylation in dapagliflozin-treated db/db mice, a type 2 diabetic model. After 8-week treatment of dapagliflozin for db/db mice having a proximal tubule-specific tdTomato reporter, tdTomato-positive cells were isolated by FACS. Pathway analysis of RNA sequencing of isolated tubular epithelia revealed that oxidative phosphorylation was downregulated in dapagliflozin-treated mice. However, depletion of renal tissue ATP content in db/db mice was ameliorated by dapagliflozin administration. Pimonidazole staining demonstrated renal cortical tissue hypoxia in db/db mice, which was improved by dapagliflozin administration. This study suggests that dapagliflozin can ameliorate the excessive oxygen and ATP consumption, and subsequent tissue hypoxia in the diabetic kidney, which may explain, in part, the responsible mechanisms of the renoprotective effects of dapagliflozin. Oxidative phosphorylation is downregulated by dapagliflozin in the proximal tubules of diabetic kidneys. Renal tissue ATP content increased in dapagliflozin-treated diabetic mice. Dapagliflozin improved renal cortical hypoxia in type 2 diabetic mice.
Collapse
Affiliation(s)
- Noriko Uehara-Watanabe
- Department of Nephrology, Graduate School of Medical Science, Kyoto Prefectural University of Medicine, Kyoto, Japan
| | - Natsuko Okuno-Ozeki
- Department of Nephrology, Graduate School of Medical Science, Kyoto Prefectural University of Medicine, Kyoto, Japan
| | - Itaru Nakamura
- Department of Nephrology, Graduate School of Medical Science, Kyoto Prefectural University of Medicine, Kyoto, Japan
| | - Tomohiro Nakata
- Department of Nephrology, Graduate School of Medical Science, Kyoto Prefectural University of Medicine, Kyoto, Japan
| | - Kunihiro Nakai
- Department of Nephrology, Graduate School of Medical Science, Kyoto Prefectural University of Medicine, Kyoto, Japan
| | - Aya Yagi-Tomita
- Department of Nephrology, Graduate School of Medical Science, Kyoto Prefectural University of Medicine, Kyoto, Japan
| | - Tomoharu Ida
- Department of Nephrology, Graduate School of Medical Science, Kyoto Prefectural University of Medicine, Kyoto, Japan
| | - Noriyuki Yamashita
- Department of Nephrology, Graduate School of Medical Science, Kyoto Prefectural University of Medicine, Kyoto, Japan
| | - Michitsugu Kamezaki
- Department of Nephrology, Graduate School of Medical Science, Kyoto Prefectural University of Medicine, Kyoto, Japan
| | - Yuhei Kirita
- Department of Nephrology, Graduate School of Medical Science, Kyoto Prefectural University of Medicine, Kyoto, Japan
| | - Satoaki Matoba
- Department of Cardiovascular Medicine, Graduate School of Medical Science, Kyoto Prefectural University of Medicine, Kyoto, Japan
| | - Keiichi Tamagaki
- Department of Nephrology, Graduate School of Medical Science, Kyoto Prefectural University of Medicine, Kyoto, Japan
| | - Tetsuro Kusaba
- Department of Nephrology, Graduate School of Medical Science, Kyoto Prefectural University of Medicine, Kyoto, Japan
| |
Collapse
|
13
|
Siddiqui I, Bilkey J, McKee TD, Serra S, Pintilie M, Do T, Xu J, Tsao MS, Gallinger S, Hill RP, Hedley DW, Dhani NC. Digital quantitative tissue image analysis of hypoxia in resected pancreatic ductal adenocarcinomas. Front Oncol 2022; 12:926497. [PMID: 35978831 PMCID: PMC9376475 DOI: 10.3389/fonc.2022.926497] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2022] [Accepted: 06/30/2022] [Indexed: 11/13/2022] Open
Abstract
BackgroundTumor hypoxia is theorized to contribute to the aggressive biology of pancreatic ductal adenocarcinoma (PDAC). We previously reported that hypoxia correlated with rapid tumor growth and metastasis in patient-derived xenografts. Anticipating a prognostic relevance of hypoxia in patient tumors, we developed protocols for automated semi-quantitative image analysis to provide an objective, observer-independent measure of hypoxia. We further validated this method which can reproducibly estimate pimonidazole-detectable hypoxia in a high-through put manner.MethodsWe studied the performance of three automated image analysis platforms in scoring pimonidazole-detectable hypoxia in resected PDAC (n = 10) in a cohort of patients enrolled in PIMO-PANC. Multiple stained tumor sections were analyzed on three independent image-analysis platforms, Aperio Genie (AG), Definiens Tissue Studio (TS), and Definiens Developer (DD), which comprised of a customized rule set.ResultsThe output from Aperio Genie (AG) had good concordance with manual scoring, but the workflow was resource-intensive and not suited for high-throughput analysis. TS analysis had high levels of variability related to misclassification of cells class, while the customized rule set of DD had a high level of reliability with an intraclass coefficient of more than 85%.DiscussionThis work demonstrates the feasibility of developing a robust, high-performance pipeline for an automated, quantitative scoring of pimonidazole-detectable hypoxia in patient tumors.
Collapse
Affiliation(s)
- Iram Siddiqui
- Department of Pediatric Laboratory Medicine, The Hospital for Sick Children, Toronto, ON, Canada
- *Correspondence: Iram Siddiqui,
| | - Jade Bilkey
- Spatio-temporal Targeting and Amplification of Radiation Response (STTARR), University Health Network, Toronto, ON, Canada
| | - Trevor D. McKee
- Spatio-temporal Targeting and Amplification of Radiation Response (STTARR), University Health Network, Toronto, ON, Canada
| | - Stefano Serra
- Department of Pathology, Toronto General Hospital, Toronto, ON, Canada
| | - Melania Pintilie
- Department of Biostatistics, The Princess Margaret Cancer Centre, Toronto, ON, Canada
| | - Trevor Do
- Spatio-temporal Targeting and Amplification of Radiation Response (STTARR), University Health Network, Toronto, ON, Canada
| | - Jing Xu
- Department of Medical Oncology, The Princess Margaret Cancer Centre, Toronto, ON, Canada
| | - Ming-Sound Tsao
- Department of Pathology, Toronto General Hospital, Toronto, ON, Canada
| | - Steve Gallinger
- PanCuRx Translational Research Initiative, Ontario Institute for Cancer Research, Toronto, ON, Canada
- Hepato-Pancreatico-Biliary Surgical Oncology Program, University Health Network, Toronto, ON, Canada
| | - Richard P. Hill
- Medicine Program, The Princess Margaret Cancer Centre/Ontario Cancer Institute, Radiation Toronto, ON, Canada
| | - David W. Hedley
- Department of Medical Oncology, The Princess Margaret Cancer Centre, Toronto, ON, Canada
| | - Neesha C. Dhani
- Department of Medical Oncology, The Princess Margaret Cancer Centre, Toronto, ON, Canada
| |
Collapse
|
14
|
Woods PS, Kimmig LM, Sun KA, Meliton AY, Shamaa OR, Tian Y, Cetin-Atalay R, Sharp WW, Hamanaka RB, Mutlu GM. HIF-1α induces glycolytic reprograming in tissue-resident alveolar macrophages to promote cell survival during acute lung injury. eLife 2022; 11:e77457. [PMID: 35822617 PMCID: PMC9323005 DOI: 10.7554/elife.77457] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2022] [Accepted: 07/10/2022] [Indexed: 12/03/2022] Open
Abstract
Cellular metabolism is a critical regulator of macrophage effector function. Tissue-resident alveolar macrophages (TR-AMs) inhabit a unique niche marked by high oxygen and low glucose. We have recently shown that in contrast to bone marrow-derived macrophages (BMDMs), TR-AMs do not utilize glycolysis and instead predominantly rely on mitochondrial function for their effector response. It is not known how changes in local oxygen concentration that occur during conditions such as acute respiratory distress syndrome (ARDS) might affect TR-AM metabolism and function; however, ARDS is associated with progressive loss of TR-AMs, which correlates with the severity of disease and mortality. Here, we demonstrate that hypoxia robustly stabilizes HIF-1α in TR-AMs to promote a glycolytic phenotype. Hypoxia altered TR-AM metabolite signatures, cytokine production, and decreased their sensitivity to the inhibition of mitochondrial function. By contrast, hypoxia had minimal effects on BMDM metabolism. The effects of hypoxia on TR-AMs were mimicked by FG-4592, a HIF-1α stabilizer. Treatment with FG-4592 decreased TR-AM death and attenuated acute lung injury in mice. These findings reveal the importance of microenvironment in determining macrophage metabolic phenotype and highlight the therapeutic potential in targeting cellular metabolism to improve outcomes in diseases characterized by acute inflammation.
Collapse
Affiliation(s)
- Parker S Woods
- Department of Medicine, Section of Pulmonary and Critical Care Medicine, The University of ChicagoChicagoUnited States
| | - Lucas M Kimmig
- Department of Medicine, Section of Pulmonary and Critical Care Medicine, The University of ChicagoChicagoUnited States
| | - Kaitlyn A Sun
- Department of Medicine, Section of Pulmonary and Critical Care Medicine, The University of ChicagoChicagoUnited States
| | - Angelo Y Meliton
- Department of Medicine, Section of Pulmonary and Critical Care Medicine, The University of ChicagoChicagoUnited States
| | - Obada R Shamaa
- Department of Medicine, Section of Pulmonary and Critical Care Medicine, The University of ChicagoChicagoUnited States
| | - Yufeng Tian
- Department of Medicine, Section of Pulmonary and Critical Care Medicine, The University of ChicagoChicagoUnited States
| | - Rengül Cetin-Atalay
- Department of Medicine, Section of Pulmonary and Critical Care Medicine, The University of ChicagoChicagoUnited States
| | - Willard W Sharp
- Department of Medicine, Section of Emergency Medicine, The University of ChicagoChicagoUnited States
| | - Robert B Hamanaka
- Department of Medicine, Section of Pulmonary and Critical Care Medicine, The University of ChicagoChicagoUnited States
| | - Gökhan M Mutlu
- Department of Medicine, Section of Pulmonary and Critical Care Medicine, The University of ChicagoChicagoUnited States
| |
Collapse
|
15
|
Higgins GS, Hammond EM. Elucidating the role of transiently hypoxic tumour cells on radiation resistance. Br J Cancer 2022; 126:971-972. [PMID: 35197582 PMCID: PMC8980025 DOI: 10.1038/s41416-022-01741-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2021] [Revised: 01/11/2022] [Accepted: 02/03/2022] [Indexed: 11/09/2022] Open
Abstract
The link between hypoxic conditions and radiation sensitivity is well-established, however the dynamic nature of hypoxia is often overlooked. The contribution of acute/transient hypoxia versus chronic conditions to radiosensitivity has been investigated by Wadsworth et al. using two hypoxia markers and pentoxifylline to increase blood flow to regions of transient hypoxia.
Collapse
Affiliation(s)
- Geoff S Higgins
- Oxford Institute for Radiation Oncology, Department of Oncology, The University of Oxford, Oxford, OX3 7DQ, UK
| | - Ester M Hammond
- Oxford Institute for Radiation Oncology, Department of Oncology, The University of Oxford, Oxford, OX3 7DQ, UK.
| |
Collapse
|
16
|
Chang WL, Su YH. Zygotic hypoxia-inducible factor alpha regulates spicule elongation in the sea urchin embryo. Dev Biol 2022; 484:63-74. [DOI: 10.1016/j.ydbio.2022.02.004] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2021] [Revised: 01/28/2022] [Accepted: 02/09/2022] [Indexed: 12/15/2022]
|
17
|
Assessment of hypoxia by pimonidazole staining following radiotherapy. Methods Cell Biol 2022; 172:179-189. [DOI: 10.1016/bs.mcb.2022.07.002] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022]
|
18
|
Immunofluorescence-Based Method to Assess Cancer Biomarker in the Hypoxic Region of the Tumor. Methods Mol Biol 2022; 2413:37-43. [PMID: 35044652 PMCID: PMC9116888 DOI: 10.1007/978-1-0716-1896-7_5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/03/2023]
Abstract
The development of novel imaging technologies allows the analysis of the expression and spatial distribution of multiple markers simultaneously, providing necessary information about a cellular identity and the surrounding microenvironment. This chapter describes the utilization of immunofluorescence to identify such biomarkers in fixed tissue from prostate cancer (PCa) xenografts. One such marker detectable by immunofluorescence is pimonidazole, which has been utilized to locate areas of low oxygen (hypoxia). Pimonidazole, in combination with other biomarkers, could be utilized to identify "niches" in the microenvironment harboring more aggressive cells both within and outside hypoxic areas. Specifically, we describe the method to use pimonidazole for the identification of hypoxic regions in PCa xenograft tumors along with CPT1A (carnitine palmitoyltransferase 1A) expression, an indicator of β-oxidation. This approach could be useful to characterize various biomarkers in the complex hypoxic tumor microenvironment.
Collapse
|
19
|
Rios-Colon L, Kumar P, Kim S, Sharma M, Su Y, Kumar A, Singh S, Stocks N, Liu L, Joshi M, Schlaepfer IR, Kumar D, Deep G. Carnitine Palmitoyltransferase 1 Regulates Prostate Cancer Growth under Hypoxia. Cancers (Basel) 2021; 13:cancers13246302. [PMID: 34944922 PMCID: PMC8699124 DOI: 10.3390/cancers13246302] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2021] [Accepted: 12/07/2021] [Indexed: 12/19/2022] Open
Abstract
Simple Summary Cancer cell survival in hypoxia areas, with low oxygen and food supply as well as abundant waste material, is critical to their aggressiveness and associated with disease relapse and mortality. Therefore, it is vital to understand the molecular regulators of cancer cell survival under these harsh physiological conditions. In the present study, we assessed the role of a mitochondrial protein carnitine palmitoyltransferase (CPT1A) in regulating prostate cancer (PCa) cell survival and proliferation under hypoxic conditions in both cell culture and animal models. The results showed that CPT1A expression in PCa cells is key to their survival and proliferation in the hypoxic tumor microenvironment. These results have high translational significance in improving cancer prognosis and therapy. Abstract Hypoxia and hypoxia-related biomarkers are the major determinants of prostate cancer (PCa) aggressiveness. Therefore, a better understanding of molecular players involved in PCa cell survival under hypoxia could offer novel therapeutic targets. We previously reported a central role of mitochondrial protein carnitine palmitoyltransferase (CPT1A) in PCa progression, but its role in regulating PCa survival under hypoxia remains unknown. Here, we employed PCa cells (22Rv1 and MDA-PCa-2b) with knockdown or overexpression of CPT1A and assessed their survival under hypoxia, both in cell culture and in vivo models. The results showed that CPT1A knockdown in PCa cells significantly reduced their viability, clonogenicity, and sphere formation under hypoxia, while its overexpression increased their proliferation, clonogenicity, and sphere formation. In nude mice, 22Rv1 xenografts with CPT1A knockdown grew significantly slower compared to vector control cells (~59% reduction in tumor volume at day 29). On the contrary, CPT1A-overexpressing 22Rv1 xenografts showed higher tumor growth compared to vector control cells (~58% higher tumor volume at day 40). Pathological analyses revealed lesser necrotic areas in CPT1A knockdown tumors and higher necrotic areas in CPT1A overexpressing tumors. Immunofluorescence analysis of tumors showed that CPT1A knockdown strongly compromised the hypoxic areas (pimonidazole+), while CPT1A overexpression resulted in more hypoxia areas with strong expression of proliferation biomarkers (Ki67 and cyclin D1). Finally, IHC analysis of tumors revealed a significant decrease in VEGF or VEGF-D expression but without significant changes in biomarkers associated with microvessel density. These results suggest that CPT1A regulates PCa survival in hypoxic conditions and might contribute to their aggressiveness.
Collapse
Affiliation(s)
- Leslimar Rios-Colon
- Department of Cancer Biology, Wake Forest Baptist Medical Center, Winston-Salem, NC 27157, USA; (L.R.-C.); (P.K.); (S.K.); (M.S.); (Y.S.); (A.K.); (S.S.); (N.S.); (L.L.)
- Julius L. Chambers Biomedical Biotechnology Research Institute, North Carolina Central University, Durham, NC 27707, USA;
| | - Pawan Kumar
- Department of Cancer Biology, Wake Forest Baptist Medical Center, Winston-Salem, NC 27157, USA; (L.R.-C.); (P.K.); (S.K.); (M.S.); (Y.S.); (A.K.); (S.S.); (N.S.); (L.L.)
- Division of Pathology, ICAR—Indian Veterinary Research Institute, Izatnagar 243122, India
| | - Susy Kim
- Department of Cancer Biology, Wake Forest Baptist Medical Center, Winston-Salem, NC 27157, USA; (L.R.-C.); (P.K.); (S.K.); (M.S.); (Y.S.); (A.K.); (S.S.); (N.S.); (L.L.)
| | - Mitu Sharma
- Department of Cancer Biology, Wake Forest Baptist Medical Center, Winston-Salem, NC 27157, USA; (L.R.-C.); (P.K.); (S.K.); (M.S.); (Y.S.); (A.K.); (S.S.); (N.S.); (L.L.)
| | - Yixin Su
- Department of Cancer Biology, Wake Forest Baptist Medical Center, Winston-Salem, NC 27157, USA; (L.R.-C.); (P.K.); (S.K.); (M.S.); (Y.S.); (A.K.); (S.S.); (N.S.); (L.L.)
| | - Ashish Kumar
- Department of Cancer Biology, Wake Forest Baptist Medical Center, Winston-Salem, NC 27157, USA; (L.R.-C.); (P.K.); (S.K.); (M.S.); (Y.S.); (A.K.); (S.S.); (N.S.); (L.L.)
| | - Sangeeta Singh
- Department of Cancer Biology, Wake Forest Baptist Medical Center, Winston-Salem, NC 27157, USA; (L.R.-C.); (P.K.); (S.K.); (M.S.); (Y.S.); (A.K.); (S.S.); (N.S.); (L.L.)
| | - Nalexus Stocks
- Department of Cancer Biology, Wake Forest Baptist Medical Center, Winston-Salem, NC 27157, USA; (L.R.-C.); (P.K.); (S.K.); (M.S.); (Y.S.); (A.K.); (S.S.); (N.S.); (L.L.)
| | - Liang Liu
- Department of Cancer Biology, Wake Forest Baptist Medical Center, Winston-Salem, NC 27157, USA; (L.R.-C.); (P.K.); (S.K.); (M.S.); (Y.S.); (A.K.); (S.S.); (N.S.); (L.L.)
- Center for Cancer Genomics and Precision Oncology, Wake Forest Baptist Medical Center, Winston-Salem, NC 27157, USA
- Wake Forest Baptist Comprehensive Cancer Center, Wake Forest Baptist Medical Center, Winston-Salem, NC 27157, USA
| | - Molishree Joshi
- Functional Genomics Facility, University of Colorado Anschutz Medical Campus, Aurora, CO 80045, USA;
| | - Isabel R. Schlaepfer
- Division of Medical Oncology, University of Colorado Anschutz Medical Center, Aurora, CO 80045, USA;
| | - Deepak Kumar
- Julius L. Chambers Biomedical Biotechnology Research Institute, North Carolina Central University, Durham, NC 27707, USA;
| | - Gagan Deep
- Department of Cancer Biology, Wake Forest Baptist Medical Center, Winston-Salem, NC 27157, USA; (L.R.-C.); (P.K.); (S.K.); (M.S.); (Y.S.); (A.K.); (S.S.); (N.S.); (L.L.)
- Wake Forest Baptist Comprehensive Cancer Center, Wake Forest Baptist Medical Center, Winston-Salem, NC 27157, USA
- Correspondence: ; Tel.: +336-716-9363
| |
Collapse
|
20
|
Weiss A, Lopez CA, Beavers WN, Rodriguez J, Skaar EP. Clostridioides difficile strain-dependent and strain-independent adaptations to a microaerobic environment. Microb Genom 2021; 7:000738. [PMID: 34908523 PMCID: PMC8767335 DOI: 10.1099/mgen.0.000738] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2021] [Accepted: 11/02/2021] [Indexed: 12/18/2022] Open
Abstract
Clostridioides difficile (formerly Clostridium difficile) colonizes the gastrointestinal tract following disruption of the microbiota and can initiate a spectrum of clinical manifestations ranging from asymptomatic to life-threatening colitis. Following antibiotic treatment, luminal oxygen concentrations increase, exposing gut microbes to potentially toxic reactive oxygen species. Though typically regarded as a strict anaerobe, C. difficile can grow at low oxygen concentrations. How this bacterium adapts to a microaerobic environment and whether those responses to oxygen are conserved amongst strains is not entirely understood. Here, two C. difficile strains (630 and CD196) were cultured in 1.5% oxygen and the transcriptional response to long-term oxygen exposure was evaluated via RNA-sequencing. During growth in a microaerobic environment, several genes predicted to protect against oxidative stress were upregulated, including those for rubrerythrins and rubredoxins. Transcription of genes involved in metal homeostasis was also positively correlated with increased oxygen levels and these genes were amongst the most differentially transcribed. To directly compare the transcriptional landscape between C. difficile strains, a 'consensus-genome' was generated. On the basis of the identified conserved genes, basal transcriptional differences as well as variations in the response to oxygen were evaluated. While several responses were similar between the strains, there were significant differences in the abundance of transcripts involved in amino acid and carbohydrate metabolism. Furthermore, intracellular metal concentrations significantly varied both in an oxygen-dependent and oxygen-independent manner. Overall, these results indicate that C. difficile adapts to grow in a low oxygen environment through transcriptional changes, though the specific strategy employed varies between strains.
Collapse
Affiliation(s)
- Andy Weiss
- Department of Pathology, Microbiology, and Immunology, Vanderbilt University Medical Center, Nashville, TN, USA
- Vanderbilt Institute for Infection, Immunology, and Inflammation, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Christopher A. Lopez
- Department of Pathology, Microbiology, and Immunology, Vanderbilt University Medical Center, Nashville, TN, USA
- Vanderbilt Institute for Infection, Immunology, and Inflammation, Vanderbilt University Medical Center, Nashville, TN, USA
- Department of Biological Sciences, California State University Sacramento, Sacramento, CA, USA
| | - William N. Beavers
- Department of Pathology, Microbiology, and Immunology, Vanderbilt University Medical Center, Nashville, TN, USA
- Vanderbilt Institute for Infection, Immunology, and Inflammation, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Jhoana Rodriguez
- Department of Biological Sciences, California State University Sacramento, Sacramento, CA, USA
| | - Eric P. Skaar
- Department of Pathology, Microbiology, and Immunology, Vanderbilt University Medical Center, Nashville, TN, USA
- Vanderbilt Institute for Infection, Immunology, and Inflammation, Vanderbilt University Medical Center, Nashville, TN, USA
| |
Collapse
|
21
|
Liapis E, Karlas A, Klemm U, Ntziachristos V. Chemotherapeutic effects on breast tumor hemodynamics revealed by eigenspectra multispectral optoacoustic tomography (eMSOT). Theranostics 2021; 11:7813-7828. [PMID: 34335966 PMCID: PMC8315054 DOI: 10.7150/thno.56173] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2020] [Accepted: 03/24/2021] [Indexed: 12/11/2022] Open
Abstract
Non-invasive monitoring of hemodynamic tumor responses to chemotherapy could provide unique insights into the development of therapeutic resistance and inform therapeutic decision-making in the clinic. Methods: Here, we examined the longitudinal and dynamic effects of the common chemotherapeutic drug Taxotere on breast tumor (KPL-4) blood volume and oxygen saturation using eigenspectra multispectral optoacoustic tomography (eMSOT) imaging over a period of 41 days. Tumor vascular function was assessed by dynamic oxygen-enhanced eMSOT (OE-eMSOT). The obtained in vivo optoacoustic data were thoroughly validated by ex vivo cryoimaging and immunohistochemical staining against markers of vascularity and hypoxia. Results: We provide the first preclinical evidence that prolonged treatment with Taxotere causes a significant drop in mean whole tumor oxygenation. Furthermore, application of OE-eMSOT showed a diminished vascular response in Taxotere-treated tumors and revealed the presence of static blood pools, indicating increased vascular permeability. Conclusion: Our work has important translational implications and supports the feasibility of eMSOT imaging for non-invasive assessment of tumor microenvironmental responses to chemotherapy.
Collapse
|
22
|
Hare GMT, Zhang Y, Chin K, Thai K, Jacobs E, Cazorla‐Bak MP, Nghiem L, Wilson DF, Vinogradov SA, Connelly KA, Mazer CD, Evans RG, Gilbert RE. Impact of sodium glucose linked cotransporter-2 inhibition on renal microvascular oxygen tension in a rodent model of diabetes mellitus. Physiol Rep 2021; 9:e14890. [PMID: 34184431 PMCID: PMC8239445 DOI: 10.14814/phy2.14890] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2021] [Revised: 06/02/2021] [Accepted: 05/04/2021] [Indexed: 01/10/2023] Open
Abstract
BACKGROUND The mechanisms whereby inhibitors of sodium-glucose linked cotransporter-2 (SGLT2) exert their nephroprotective effects in patients with diabetes are incompletely understood but have been hypothesized to include improved tissue oxygen tension within the renal cortex. The impact of SGLT2 inhibition is likely complex and region specific within the kidney. We hypothesize that SGLT2 inhibitors have differential effects on renal tissue oxygen delivery and consumption in specific regions of the diabetic kidney, including the superficial cortex, containing SGLT2-rich components of proximal tubules, versus the deeper cortex and outer medulla, containing predominantly SGLT1 receptors. METHODS We measured glomerular filtration rate (GFR), microvascular kidney oxygen tension (Pk O2 ), erythropoietin (EPO) mRNA, and reticulocyte count in diabetic rats (streptozotocin) treated with the SGLT2 inhibitor, dapagliflozin. Utilizing phosphorescence quenching by oxygen and an intravascular oxygen sensitive probe (Oxyphor PdG4); we explored the effects of SGLT2 inhibition on Pk O2 in a region-specific manner, in vivo, in diabetic and non-diabetic rats. Superficial renal cortical or deeper cortical and outer medullary Pk O2 were measured utilizing excitations with blue and red light wavelengths, respectively. RESULTS In diabetic rats treated with dapagliflozin, measurement within the superficial cortex (blue light) demonstrated no change in Pk O2 . By contrast, measurements in the deeper cortex and outer medulla (red light) demonstrated a significant reduction in Pk O2 in dapagliflozin treated diabetic rats (p = 0.014). Consistent with these findings, GFR was decreased, hypoxia-responsive EPO mRNA levels were elevated and reticulocyte counts were increased with SGLT2 inhibition in diabetic rats (p < 0.05 for all). CONCLUSIONS These findings indicate that microvascular kidney oxygen tension is maintained in the superficial cortex but reduced in deeper cortical and outer medullary tissue, possibly due to the regional impact of SGLT-2 inhibition on tissue metabolism. This reduction in deeper Pk O2 had biological impact as demonstrated by increased renal EPO mRNA levels and circulating reticulocyte count.
Collapse
Affiliation(s)
- Gregory M. T. Hare
- Department of AnesthesiaSt. Michael's HospitalUniversity of TorontoTorontoONCanada
- Department of PhysiologyUniversity of TorontoTorontoONCanada
- Keenan Research Centre for Biomedical Science in the Li Ka Shing Knowledge InstituteSt. Michael's HospitalTorontoONCanada
| | - Yanling Zhang
- Keenan Research Centre for Biomedical Science in the Li Ka Shing Knowledge InstituteSt. Michael's HospitalTorontoONCanada
| | - Kyle Chin
- Department of AnesthesiaSt. Michael's HospitalUniversity of TorontoTorontoONCanada
- Department of PhysiologyUniversity of TorontoTorontoONCanada
| | - Kerri Thai
- Keenan Research Centre for Biomedical Science in the Li Ka Shing Knowledge InstituteSt. Michael's HospitalTorontoONCanada
| | - Evelyn Jacobs
- Department of AnesthesiaSt. Michael's HospitalUniversity of TorontoTorontoONCanada
| | - Melina P. Cazorla‐Bak
- Department of AnesthesiaSt. Michael's HospitalUniversity of TorontoTorontoONCanada
- Department of PhysiologyUniversity of TorontoTorontoONCanada
| | - Linda Nghiem
- Keenan Research Centre for Biomedical Science in the Li Ka Shing Knowledge InstituteSt. Michael's HospitalTorontoONCanada
| | - David F. Wilson
- Department of Biochemistry and BiophysicsSchool of MedicineUniversity of PennsylvaniaPhiladelphiaPAUSA
| | - Sergei A. Vinogradov
- Department of Biochemistry and BiophysicsSchool of MedicineUniversity of PennsylvaniaPhiladelphiaPAUSA
| | - Kim A. Connelly
- Department of PhysiologyUniversity of TorontoTorontoONCanada
- Keenan Research Centre for Biomedical Science in the Li Ka Shing Knowledge InstituteSt. Michael's HospitalTorontoONCanada
- Department of MedicineDivision of CardiologySt. Michael's HospitalUniversity of TorontoTorontoONCanada
| | - C. David Mazer
- Department of AnesthesiaSt. Michael's HospitalUniversity of TorontoTorontoONCanada
- Department of PhysiologyUniversity of TorontoTorontoONCanada
- Keenan Research Centre for Biomedical Science in the Li Ka Shing Knowledge InstituteSt. Michael's HospitalTorontoONCanada
- Institute of Medical ScienceUniversity of TorontoTorontoONCanada
| | - Roger G. Evans
- Cardiovascular Disease ProgramBiomedicine Discovery Institute and Department of PhysiologyMonash UniversityMelbourneVicAustralia
| | - Richard E. Gilbert
- Keenan Research Centre for Biomedical Science in the Li Ka Shing Knowledge InstituteSt. Michael's HospitalTorontoONCanada
- Department of MedicineDivision of EndocrinologySt. Michael's HospitalUniversity of TorontoTorontoONCanada
| |
Collapse
|
23
|
Lin JH, Yu YW, Chuang YC, Lee CH, Chen CC. ATF3-Expressing Large-Diameter Sensory Afferents at Acute Stage as Bio-Signatures of Persistent Pain Associated with Lumbar Radiculopathy. Cells 2021; 10:cells10050992. [PMID: 33922541 PMCID: PMC8145235 DOI: 10.3390/cells10050992] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2021] [Revised: 04/09/2021] [Accepted: 04/21/2021] [Indexed: 11/16/2022] Open
Abstract
The mechanism of pain chronicity is largely unknown in lumbar radiculopathy (LR). The anatomical location of nerve injury is one of the important factors associated with pain chronicity of LR. Accumulating evidence has shown constriction distal to the dorsal root ganglion (DRG) caused more severe radiculopathy than constriction proximal to the DRG; thereby, the mechanism of pain chronicity in LR could be revealed by comparing the differences in pathological changes of DRGs between nerve constriction distal and proximal to the DRG. Here, we used 2 rat models of LR with nerve constriction distal or proximal to the DRG to probe how the different nerve injury sites could differentially affect pain chronicity and the pathological changes of DRG neuron subpopulations. As expected, rats with nerve constriction distal to the DRG showed more persistent pain behaviors than those with nerve constriction proximal to the DRG in 50% paw withdraw threshold, weight-bearing test, and acetone test. One day after the operation, distal and proximal nerve constriction showed differential pathological changes of DRG. The ratios of activating transcription factor3 (ATF3)-positive DRG neurons were significantly higher in rats with nerve constriction distal to DRG than those with nerve constriction proximal to DRG. In subpopulation analysis, the ratios of ATF3-immunoreactivity (IR) in neurofilament heavy chain (NFH)-positive DRG neurons significantly increased in distal nerve constriction compared to proximal nerve constriction; although, both distal and proximal nerve constriction presented increased ratios of ATF3-IR in calcitonin gene-related peptide (CGRP)-positive DRG neurons. Moreover, the nerve constriction proximal to DRG caused more hypoxia than did that distal to DRG. Together, ATF3 expression in NHF-positive DRG neurons at the acute stage is a potential bio-signature of persistent pain in rat models of LR.
Collapse
Affiliation(s)
- Jiann-Her Lin
- Department of Neurosurgery, Taipei Medical University Hospital, Taipei 110301, Taiwan;
- Department of Surgery, Division of Neurosurgery, School of Medicine, College of Medicine, Taipei Medical University, Taipei 110301, Taiwan;
- Taipei Neuroscience Institute, Taipei Medical University, Taipei 110301, Taiwan
- Institute of Biomedical Sciences, Academia Sinica, Taipei 115201, Taiwan; (Y.-C.C.); (C.-H.L.)
| | - Yu-Wen Yu
- Department of Surgery, Division of Neurosurgery, School of Medicine, College of Medicine, Taipei Medical University, Taipei 110301, Taiwan;
- Institute of Biomedical Sciences, Academia Sinica, Taipei 115201, Taiwan; (Y.-C.C.); (C.-H.L.)
| | - Yu-Chia Chuang
- Institute of Biomedical Sciences, Academia Sinica, Taipei 115201, Taiwan; (Y.-C.C.); (C.-H.L.)
| | - Cheng-Han Lee
- Institute of Biomedical Sciences, Academia Sinica, Taipei 115201, Taiwan; (Y.-C.C.); (C.-H.L.)
| | - Chih-Cheng Chen
- Institute of Biomedical Sciences, Academia Sinica, Taipei 115201, Taiwan; (Y.-C.C.); (C.-H.L.)
- Neuroscience Program of Academia Sinica, Academia Sinica, Taipei 115201, Taiwan
- Taiwan Mouse Clinic, Biomedical Translation Research Center, Academia Sinica, Taipei 115202, Taiwan
- Correspondence:
| |
Collapse
|
24
|
Assessment of tumor hypoxia and perfusion in recurrent glioblastoma following bevacizumab failure using MRI and 18F-FMISO PET. Sci Rep 2021; 11:7632. [PMID: 33828310 PMCID: PMC8027395 DOI: 10.1038/s41598-021-84331-5] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2020] [Accepted: 02/03/2021] [Indexed: 01/16/2023] Open
Abstract
Tumoral hypoxia correlates with worse outcomes in glioblastoma (GBM). While bevacizumab is routinely used to treat recurrent GBM, it may exacerbate hypoxia. Evofosfamide is a hypoxia-targeting prodrug being tested for recurrent GBM. To characterize resistance to bevacizumab and identify those with recurrent GBM who may benefit from evofosfamide, we ascertained MRI features and hypoxia in patients with GBM progression receiving both agents. Thirty-three patients with recurrent GBM refractory to bevacizumab were enrolled. Patients underwent MR and 18F-FMISO PET imaging at baseline and 28 days. Tumor volumes were determined, MRI and 18F-FMISO PET-derived parameters calculated, and Spearman correlations between parameters assessed. Progression-free survival decreased significantly with hypoxic volume [hazard ratio (HR) = 1.67, 95% confidence interval (CI) 1.14 to 2.46, P = 0.009] and increased significantly with time to the maximum value of the residue (Tmax) (HR = 0.54, 95% CI 0.34 to 0.88, P = 0.01). Overall survival decreased significantly with hypoxic volume (HR = 1.71, 95% CI 1.12 to 12.61, p = 0.01), standardized relative cerebral blood volume (srCBV) (HR = 1.61, 95% CI 1.09 to 2.38, p = 0.02), and increased significantly with Tmax (HR = 0.31, 95% CI 0.15 to 0.62, p < 0.001). Decreases in hypoxic volume correlated with longer overall and progression-free survival, and increases correlated with shorter overall and progression-free survival. Hypoxic volume and volume ratio were positively correlated (rs = 0.77, P < 0.0001), as were hypoxia volume and T1 enhancing tumor volume (rs = 0.75, P < 0.0001). Hypoxia is a key biomarker in patients with bevacizumab-refractory GBM. Hypoxia and srCBV were inversely correlated with patient outcomes. These radiographic features may be useful in evaluating treatment and guiding treatment considerations.
Collapse
|
25
|
Hegde A, Jayaprakash P, Couillault CA, Piha-Paul S, Karp D, Rodon J, Pant S, Fu S, Dumbrava EE, Yap TA, Subbiah V, Bhosale P, Coarfa C, Higgins JP, Williams ET, Wilson TF, Lim J, Meric-Bernstam F, Sumner E, Zain H, Nguyen D, Nguyen LM, Rajapakshe K, Curran MA, Hong DS. A Phase I Dose-Escalation Study to Evaluate the Safety and Tolerability of Evofosfamide in Combination with Ipilimumab in Advanced Solid Malignancies. Clin Cancer Res 2021; 27:3050-3060. [PMID: 33771853 DOI: 10.1158/1078-0432.ccr-20-4118] [Citation(s) in RCA: 28] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2020] [Revised: 12/21/2020] [Accepted: 03/22/2021] [Indexed: 01/22/2023]
Abstract
PURPOSE As hypoxia can mediate resistance to immunotherapy, we investigated the safety, tolerability, and efficacy of combining evofosfamide, a prodrug that alleviates hypoxia, with ipilimumab, an immune checkpoint inhibitor, in immunologically "cold" cancers, which are intrinsically insensitive to immunotherapy, as well as in "hot/warm" metastatic cancers that are, atypical of such cancers, resistant to immunotherapy. PATIENTS AND METHODS In a phase I, 3+3 dose-escalation trial (NCT03098160), evofosfamide (400-640 mg/m2) and ipilimumab (3 mg/kg) were administered in four 3-week cycles. The former was administered on days 1 and 8 of cycles 1-2, while the latter was administered on day 8 of cycles 1-4. Response was assessed using immune-related RECIST and retreatment was allowed, if deemed beneficial, after completion of cycle 4 or at progression. RESULTS Twenty-two patients were enrolled, of whom 21 were evaluable, encompassing castration-resistant prostate cancer (n = 11), pancreatic cancer (n = 7), immunotherapy-resistant melanoma (n = 2), and human papillomavirus-negative head and neck cancer (n = 1). Drug-related hematologic toxicities, rash, fever, nausea, vomiting, and elevation of liver enzymes were observed in > 10% of patients. The most common drug-related grade 3 adverse event was alanine aminotransferase elevation (33.3%). Two patients discontinued ipilimumab and 4 required evofosfamide deescalation due to toxicity. Of 18 patients with measurable disease at baseline, 3 (16.7%) achieved partial response and 12 (66.7%) achieved stable disease. The best responses were observed at 560 mg/m2 evofosfamide. Preexisting immune gene signatures predicted response to therapy, while hypermetabolic tumors predicted progression. Responders also showed improved peripheral T-cell proliferation and increased intratumoral T-cell infiltration into hypoxia. CONCLUSIONS No new or unexpected safety signals were observed from combining evofosfamide and ipilimumab, and evidence of therapeutic activity was noted.
Collapse
Affiliation(s)
- Aparna Hegde
- Department of Investigational Cancer Therapeutics, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Priyamvada Jayaprakash
- Department of Immunology, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Coline A Couillault
- Department of Immunology, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Sarina Piha-Paul
- Department of Investigational Cancer Therapeutics, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Daniel Karp
- Department of Investigational Cancer Therapeutics, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Jordi Rodon
- Department of Investigational Cancer Therapeutics, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Shubham Pant
- Department of Investigational Cancer Therapeutics, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Siqing Fu
- Department of Investigational Cancer Therapeutics, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Ecaterina E Dumbrava
- Department of Investigational Cancer Therapeutics, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Timothy A Yap
- Department of Investigational Cancer Therapeutics, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Vivek Subbiah
- Department of Investigational Cancer Therapeutics, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Priya Bhosale
- Department of Diagnostic Radiology, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Cristian Coarfa
- Department of Molecular and Cellular Biology, Baylor College of Medicine, Houston, Texas
| | | | | | | | - JoAnn Lim
- Department of Investigational Cancer Therapeutics, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Funda Meric-Bernstam
- Department of Investigational Cancer Therapeutics, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Elizabeth Sumner
- Department of Investigational Cancer Therapeutics, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Hira Zain
- Department of Investigational Cancer Therapeutics, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Di Nguyen
- Department of Investigational Cancer Therapeutics, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Ly M Nguyen
- Department of Investigational Cancer Therapeutics, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Kimal Rajapakshe
- Department of Molecular and Cellular Biology, Baylor College of Medicine, Houston, Texas
| | - Michael A Curran
- Department of Immunology, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - David S Hong
- Department of Investigational Cancer Therapeutics, The University of Texas MD Anderson Cancer Center, Houston, Texas.
| |
Collapse
|
26
|
Jardim-Perassi BV, Mu W, Huang S, Tomaszewski MR, Poleszczuk J, Abdalah MA, Budzevich MM, Dominguez-Viqueira W, Reed DR, Bui MM, Johnson JO, Martinez GV, Gillies RJ. Deep-learning and MR images to target hypoxic habitats with evofosfamide in preclinical models of sarcoma. Theranostics 2021; 11:5313-5329. [PMID: 33859749 PMCID: PMC8039958 DOI: 10.7150/thno.56595] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2020] [Accepted: 02/03/2021] [Indexed: 11/05/2022] Open
Abstract
Rationale: Hypoxic regions (habitats) within tumors are heterogeneously distributed and can be widely variant. Hypoxic habitats are generally pan-therapy resistant. For this reason, hypoxia-activated prodrugs (HAPs) have been developed to target these resistant volumes. The HAP evofosfamide (TH-302) has shown promise in preclinical and early clinical trials of sarcoma. However, in a phase III clinical trial of non-resectable soft tissue sarcomas, TH-302 did not improve survival in combination with doxorubicin (Dox), possibly due to a lack of patient stratification based on hypoxic status. Therefore, we used magnetic resonance imaging (MRI) to identify hypoxic habitats and non-invasively follow therapies response in sarcoma mouse models. Methods: We developed deep-learning (DL) models to identify hypoxia, using multiparametric MRI and co-registered histology, and monitored response to TH-302 in a patient-derived xenograft (PDX) of rhabdomyosarcoma and a syngeneic model of fibrosarcoma (radiation-induced fibrosarcoma, RIF-1). Results: A DL convolutional neural network showed strong correlations (>0.76) between the true hypoxia fraction in histology and the predicted hypoxia fraction in multiparametric MRI. TH-302 monotherapy or in combination with Dox delayed tumor growth and increased survival in the hypoxic PDX model (p<0.05), but not in the RIF-1 model, which had a lower volume of hypoxic habitats. Control studies showed that RIF-1 resistance was due to hypoxia and not other causes. Notably, PDX tumors developed resistance to TH-302 under prolonged treatment that was not due to a reduction in hypoxic volumes. Conclusion: Artificial intelligence analysis of pre-therapy MR images can predict hypoxia and subsequent response to HAPs. This approach can be used to monitor therapy response and adapt schedules to forestall the emergence of resistance.
Collapse
|
27
|
Nadeau JR, Arnold BM, Johnston JM, Muir GD, Verge VMK. Acute intermittent hypoxia enhances regeneration of surgically repaired peripheral nerves in a manner akin to electrical stimulation. Exp Neurol 2021; 341:113671. [PMID: 33684407 DOI: 10.1016/j.expneurol.2021.113671] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2020] [Revised: 02/16/2021] [Accepted: 02/25/2021] [Indexed: 12/12/2022]
Abstract
The intrinsic repair response of injured peripheral neurons is enhanced by brief electrical stimulation (ES) at time of surgical repair, resulting in improved regeneration in rodents and humans. However, ES is invasive. Acute intermittent hypoxia (AIH) - breathing alternate cycles of regular air and air with ~50% normal oxygen levels (11% O2), considered mild hypoxia, is an emerging, promising non-invasive therapy that promotes motor function in spinal cord injured rats and humans. AIH can increase neural activity and under moderately severe hypoxic conditions improves repair of peripherally crushed nerves in mice. Thus, we posited an AIH paradigm similar to that used clinically for spinal cord injury, will improve surgically repaired peripheral nerves akin to ES, including an impact on regeneration-associated gene (RAG) expression-a predictor of growth states. Alterations in early RAG expression were examined in adult male Lewis rats that underwent tibial nerve coaptation repair with either 2 days AIH or normoxia control treatment begun on day 2 post-repair, or 1 h ES treatment (20 Hz) at time of repair. Three days post-repair, AIH or ES treatments effected significant and parallel elevated RAG expression relative to normoxia control at the level of injured sensory and motor neuron cell bodies and proximal axon front. These parallel impacts on RAG expression were coupled with significant improvements in later indices of regeneration, namely enhanced myelination and increased numbers of newly myelinated fibers detected 20 mm distal to the tibial nerve repair site or sensory and motor neurons retrogradely labeled 28 mm distal to the repair site, both at 25 days post nerve repair; and improved return of toe spread function 5-10 weeks post-repair. Collectively, AIH mirrors many beneficial effects of ES on peripheral nerve repair outcomes. This highlights its potential for clinical translation as a non-invasive means to effect improved regeneration of injured peripheral nerves.
Collapse
Affiliation(s)
- J R Nadeau
- Department of Anatomy, Physiology and Pharmacology, University of Saskatchewan, Saskatoon, SK S7N 5E5, Canada; Biomedical Sciences, WCVM, University of Saskatchewan, 52 Campus Drive, Saskatoon, SK S7N 5B4, Canada; Cameco MS Neuroscience Research Centre, University of Saskatchewan, Saskatoon, SK S7K 0M7, Canada
| | - B M Arnold
- Biomedical Sciences, WCVM, University of Saskatchewan, 52 Campus Drive, Saskatoon, SK S7N 5B4, Canada; Cameco MS Neuroscience Research Centre, University of Saskatchewan, Saskatoon, SK S7K 0M7, Canada
| | - J M Johnston
- Department of Anatomy, Physiology and Pharmacology, University of Saskatchewan, Saskatoon, SK S7N 5E5, Canada; Cameco MS Neuroscience Research Centre, University of Saskatchewan, Saskatoon, SK S7K 0M7, Canada
| | - G D Muir
- Biomedical Sciences, WCVM, University of Saskatchewan, 52 Campus Drive, Saskatoon, SK S7N 5B4, Canada; Cameco MS Neuroscience Research Centre, University of Saskatchewan, Saskatoon, SK S7K 0M7, Canada
| | - V M K Verge
- Department of Anatomy, Physiology and Pharmacology, University of Saskatchewan, Saskatoon, SK S7N 5E5, Canada; Cameco MS Neuroscience Research Centre, University of Saskatchewan, Saskatoon, SK S7K 0M7, Canada.
| |
Collapse
|
28
|
Tumor Hypoxia as a Barrier in Cancer Therapy: Why Levels Matter. Cancers (Basel) 2021; 13:cancers13030499. [PMID: 33525508 PMCID: PMC7866096 DOI: 10.3390/cancers13030499] [Citation(s) in RCA: 79] [Impact Index Per Article: 26.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2020] [Revised: 01/22/2021] [Accepted: 01/23/2021] [Indexed: 02/07/2023] Open
Abstract
Simple Summary Hypoxia is a common feature of solid tumors and associated with poor outcome in most cancer types and treatment modalities, including radiotherapy, chemotherapy, surgery and, most likely, immunotherapy. Emerging strategies, such as proton therapy and combination therapies with radiation and hypoxia targeted drugs, provide new opportunities to overcome the hypoxia barrier and improve therapeutic outcome. Hypoxia is heterogeneously distributed both between and within tumors and shows large variations across patients not only in prevalence, but importantly, also in level. To best exploit the emerging strategies, a better understanding of how individual hypoxia levels from mild to severe affect tumor biology is vital. Here, we discuss our current knowledge on this topic and how we should proceed to gain more insight into the field. Abstract Hypoxia arises in tumor regions with insufficient oxygen supply and is a major barrier in cancer treatment. The distribution of hypoxia levels is highly heterogeneous, ranging from mild, almost non-hypoxic, to severe and anoxic levels. The individual hypoxia levels induce a variety of biological responses that impair the treatment effect. A stronger focus on hypoxia levels rather than the absence or presence of hypoxia in our investigations will help development of improved strategies to treat patients with hypoxic tumors. Current knowledge on how hypoxia levels are sensed by cancer cells and mediate cellular responses that promote treatment resistance is comprehensive. Recently, it has become evident that hypoxia also has an important, more unexplored role in the interaction between cancer cells, stroma and immune cells, influencing the composition and structure of the tumor microenvironment. Establishment of how such processes depend on the hypoxia level requires more advanced tumor models and methodology. In this review, we describe promising model systems and tools for investigations of hypoxia levels in tumors. We further present current knowledge and emerging research on cellular responses to individual levels, and discuss their impact in novel therapeutic approaches to overcome the hypoxia barrier.
Collapse
|
29
|
Abstract
Over the last few years, cancer immunotherapy experienced tremendous developments and it is nowadays considered a promising strategy against many types of cancer. However, the exclusion of lymphocytes from the tumor nest is a common phenomenon that limits the efficiency of immunotherapy in solid tumors. Despite several mechanisms proposed during the years to explain the immune excluded phenotype, at present, there is no integrated understanding about the role played by different models of immune exclusion in human cancers. Hypoxia is a hallmark of most solid tumors and, being a multifaceted and complex condition, shapes in a unique way the tumor microenvironment, affecting gene transcription and chromatin remodeling. In this review, we speculate about an upstream role for hypoxia as a common biological determinant of immune exclusion in solid tumors. We also discuss the current state of ex vivo and in vivo imaging of hypoxic determinants in relation to T cell distribution that could mechanisms of immune exclusion and discover functional-morphological tumor features that could support clinical monitoring.
Collapse
|
30
|
Guo W, Wu C. Detection of Hypoxic Regions in the Bone Microenvironment. Methods Mol Biol 2021; 2230:345-356. [PMID: 33197024 DOI: 10.1007/978-1-0716-1028-2_20] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/11/2023]
Abstract
Oxygen serves as a critical environmental factor essential for maintaining the physiological state of a tissue. Hypoxia, or low oxygen, triggers a cascade of events which allows for cells to adapt to low oxygen tensions and to facilitate oxygen delivery required to maintain tissue homeostasis. In the bone microenvironment (BME), vascular heterogeneity, poor perfusion rates of blood vessels, and high metabolic activity of hematopoietic cells result in the generation of a unique hypoxic landscape. Importantly, in this region, hypoxia and its downstream effectors are associated with establishing stem cell niches and regulating the differentiation of committed progenitors. Given the functional importance of the hypoxic bone niche, visualizing regions of hypoxia may provide valuable insights into the mechanisms that regulate tissue homeostasis. Here, we describe the utilization of the nitroimidazole derivative, pimonidazole, to detect hypoxic regions within the BME.
Collapse
Affiliation(s)
- Wendi Guo
- Department of Orthopaedic Surgery, Duke University School of Medicine, Durham, NC, USA
- Department of Pharmacology and Cancer Biology, Duke University School of Medicine, Durham, NC, USA
| | - Colleen Wu
- Department of Orthopaedic Surgery, Duke University School of Medicine, Durham, NC, USA.
- Department of Pharmacology and Cancer Biology, Duke University School of Medicine, Durham, NC, USA.
- Department of Cell Biology, Duke University School of Medicine, Durham, NC, USA.
| |
Collapse
|
31
|
Scarpelli ML, Healey DR, Fuentes A, Kodibagkar VD, Quarles CC. Correlation of Tumor Hypoxia Metrics Derived from 18F-Fluoromisonidazole Positron Emission Tomography and Pimonidazole Fluorescence Images of Optically Cleared Brain Tissue. Tomography 2020; 6:379-388. [PMID: 33364428 PMCID: PMC7744194 DOI: 10.18383/j.tom.2020.00046] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/03/2023] Open
Abstract
18F-fluoromisonidazole (FMISO) positron emission tomography (PET) is a widely used noninvasive imaging modality for assessing hypoxia. We describe the first spatial comparison of FMISO PET with an ex vivo reference standard for hypoxia across whole tumor volumes. Eighteen rats were orthotopically implanted with C6 or 9L brain tumors and made to undergo FMISO PET scanning. Whole brains were excised, sliced into 1-mm-thick sections, optically cleared, and fluorescently imaged for pimonidazole using an in vivo imaging system. FMISO maximum tumor uptake, maximum tumor-to-cerebellar uptake (TCmax), and hypoxic fraction (extracted 110 minutes after FMISO injection) were correlated with analogous metrics derived from pimonidazole fluorescence images. FMISO SUVmax was not significantly different between C6 and 9L brain tumors (P = .70), whereas FMISO TCmax and hypoxic fraction were significantly greater for C6 tumors (P < .01). FMISO TCmax was significantly correlated with the maximum tumor pimonidazole intensity (ρ = 0.76, P < .01), whereas FMISO SUVmax was not. FMISO tumor hypoxic fraction was significantly correlated with the pimonidazole-derived hypoxic fraction (ρ = 0.78, P < .01). Given that FMISO TCmax and tumor hypoxic fraction had strong correlations with the pimonidazole reference standard, these metrics may offer more reliable measures of tumor hypoxia than conventional PET uptake metrics (SUVmax). The voxel-wise correlation between FMISO uptake and pimonidazole intensity for a given tumor was strongly dependent on the tumor's TCmax (ρ = 0.81, P < .01) and hypoxic fraction (ρ = 0.85, P < .01), indicating PET measurements within individual voxels showed greater correlation with pimonidazole reference standard in tumors with greater hypoxia.
Collapse
Affiliation(s)
- Matthew L. Scarpelli
- Barrow Neuroimaging Innovation Center, Barrow Neurological Institute, St. Joseph’s Hospital and Medical Center, Phoenix, AZ; and
| | - Debbie R. Healey
- Barrow Neuroimaging Innovation Center, Barrow Neurological Institute, St. Joseph’s Hospital and Medical Center, Phoenix, AZ; and
| | - Alberto Fuentes
- Barrow Neuroimaging Innovation Center, Barrow Neurological Institute, St. Joseph’s Hospital and Medical Center, Phoenix, AZ; and
| | - Vikram D. Kodibagkar
- Barrow Neuroimaging Innovation Center, Barrow Neurological Institute, St. Joseph’s Hospital and Medical Center, Phoenix, AZ; and
| | - C. Chad Quarles
- Barrow Neuroimaging Innovation Center, Barrow Neurological Institute, St. Joseph’s Hospital and Medical Center, Phoenix, AZ; and
| |
Collapse
|
32
|
Kim JJ, Lee E, Ryu GR, Ko SH, Ahn YB, Song KH. Hypoxia Increases β-Cell Death by Activating Pancreatic Stellate Cells within the Islet. Diabetes Metab J 2020; 44:919-927. [PMID: 32431113 PMCID: PMC7801750 DOI: 10.4093/dmj.2019.0181] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/07/2019] [Accepted: 11/06/2019] [Indexed: 01/27/2023] Open
Abstract
BACKGROUND Hypoxia can occur in pancreatic islets in type 2 diabetes mellitus. Pancreatic stellate cells (PSCs) are activated during hypoxia. Here we aimed to investigate whether PSCs within the islet are also activated in hypoxia, causing β-cell injury. METHODS Islet and primary PSCs were isolated from Sprague Dawley rats, and cultured in normoxia (21% O2) or hypoxia (1% O2). The expression of α-smooth muscle actin (α-SMA), as measured by immunostaining and Western blotting, was used as a marker of PSC activation. Conditioned media (hypoxia-CM) were obtained from PSCs cultured in hypoxia. RESULTS Islets and PSCs cultured in hypoxia exhibited higher expressions of α-SMA than did those cultured in normoxia. Hypoxia increased the production of reactive oxygen species. The addition of N-acetyl-L-cysteine, an antioxidant, attenuated the hypoxia-induced PSC activation in islets and PSCs. Islets cultured in hypoxia-CM showed a decrease in cell viability and an increase in apoptosis. CONCLUSION PSCs within the islet are activated in hypoxia through oxidative stress and promote islet cell death, suggesting that hypoxia-induced PSC activation may contribute to β-cell loss in type 2 diabetes mellitus.
Collapse
Affiliation(s)
- Jong Jin Kim
- Division of Endocrinology and Metabolism, Department of Internal Medicine, College of Medicine, The Catholic University of Korea, Seoul, Korea
| | - Esder Lee
- Division of Endocrinology and Metabolism, Department of Internal Medicine, College of Medicine, The Catholic University of Korea, Seoul, Korea
| | - Gyeong Ryul Ryu
- Division of Endocrinology and Metabolism, Department of Internal Medicine, College of Medicine, The Catholic University of Korea, Seoul, Korea
| | - Seung-Hyun Ko
- Division of Endocrinology and Metabolism, Department of Internal Medicine, College of Medicine, The Catholic University of Korea, Seoul, Korea
| | - Yu-Bae Ahn
- Division of Endocrinology and Metabolism, Department of Internal Medicine, College of Medicine, The Catholic University of Korea, Seoul, Korea
| | - Ki-Ho Song
- Division of Endocrinology and Metabolism, Department of Internal Medicine, College of Medicine, The Catholic University of Korea, Seoul, Korea
| |
Collapse
|
33
|
Doctor A, Seifert V, Ullrich M, Hauser S, Pietzsch J. Three-Dimensional Cell Culture Systems in Radiopharmaceutical Cancer Research. Cancers (Basel) 2020; 12:cancers12102765. [PMID: 32993034 PMCID: PMC7600608 DOI: 10.3390/cancers12102765] [Citation(s) in RCA: 27] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2020] [Revised: 09/14/2020] [Accepted: 09/16/2020] [Indexed: 12/12/2022] Open
Abstract
In preclinical cancer research, three-dimensional (3D) cell culture systems such as multicellular spheroids and organoids are becoming increasingly important. They provide valuable information before studies on animal models begin and, in some cases, are even suitable for reducing or replacing animal experiments. Furthermore, they recapitulate microtumors, metastases, and the tumor microenvironment much better than monolayer culture systems could. Three-dimensional models show higher structural complexity and diverse cell interactions while reflecting (patho)physiological phenomena such as oxygen and nutrient gradients in the course of their growth or development. These interactions and properties are of great importance for understanding the pathophysiological importance of stromal cells and the extracellular matrix for tumor progression, treatment response, or resistance mechanisms of solid tumors. Special emphasis is placed on co-cultivation with tumor-associated cells, which further increases the predictive value of 3D models, e.g., for drug development. The aim of this overview is to shed light on selected 3D models and their advantages and disadvantages, especially from the radiopharmacist's point of view with focus on the suitability of 3D models for the radiopharmacological characterization of novel radiotracers and radiotherapeutics. Special attention is paid to pancreatic ductal adenocarcinoma (PDAC) as a predestined target for the development of new radionuclide-based theranostics.
Collapse
Affiliation(s)
- Alina Doctor
- Department of Radiopharmaceutical and Chemical Biology, Institute of Radiopharmaceutical Cancer Research, Helmholtz-Zentrum Dresden-Rossendorf, 01328 Dresden, Germany; (A.D.); (V.S.); (M.U.); (S.H.)
- School of Science, Faculty of Chemistry and Food Chemistry, Technische Universität Dresden, 01069 Dresden, Germany
| | - Verena Seifert
- Department of Radiopharmaceutical and Chemical Biology, Institute of Radiopharmaceutical Cancer Research, Helmholtz-Zentrum Dresden-Rossendorf, 01328 Dresden, Germany; (A.D.); (V.S.); (M.U.); (S.H.)
- School of Science, Faculty of Chemistry and Food Chemistry, Technische Universität Dresden, 01069 Dresden, Germany
| | - Martin Ullrich
- Department of Radiopharmaceutical and Chemical Biology, Institute of Radiopharmaceutical Cancer Research, Helmholtz-Zentrum Dresden-Rossendorf, 01328 Dresden, Germany; (A.D.); (V.S.); (M.U.); (S.H.)
| | - Sandra Hauser
- Department of Radiopharmaceutical and Chemical Biology, Institute of Radiopharmaceutical Cancer Research, Helmholtz-Zentrum Dresden-Rossendorf, 01328 Dresden, Germany; (A.D.); (V.S.); (M.U.); (S.H.)
| | - Jens Pietzsch
- Department of Radiopharmaceutical and Chemical Biology, Institute of Radiopharmaceutical Cancer Research, Helmholtz-Zentrum Dresden-Rossendorf, 01328 Dresden, Germany; (A.D.); (V.S.); (M.U.); (S.H.)
- School of Science, Faculty of Chemistry and Food Chemistry, Technische Universität Dresden, 01069 Dresden, Germany
- Correspondence: ; Tel.: +49-351-260-2622
| |
Collapse
|
34
|
Mahiddine K, Blaisdell A, Ma S, Créquer-Grandhomme A, Lowell CA, Erlebacher A. Relief of tumor hypoxia unleashes the tumoricidal potential of neutrophils. J Clin Invest 2020; 130:389-403. [PMID: 31600172 DOI: 10.1172/jci130952] [Citation(s) in RCA: 72] [Impact Index Per Article: 18.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2019] [Accepted: 10/08/2019] [Indexed: 12/29/2022] Open
Abstract
Polymorphonuclear neutrophils (PMNs) are increasingly recognized to influence solid tumor development, but why their effects are so context dependent and even frequently divergent remains poorly understood. Using an autochthonous mouse model of uterine cancer and the administration of respiratory hyperoxia as a means to improve tumor oxygenation, we provide in vivo evidence that hypoxia is a potent determinant of tumor-associated PMN phenotypes and direct PMN-tumor cell interactions. Upon relief of tumor hypoxia, PMNs were recruited less intensely to the tumor-bearing uterus, but the recruited cells much more effectively killed tumor cells, an activity our data moreover suggested was mediated via their production of NADPH oxidase-derived reactive oxygen species and MMP-9. Simultaneously, their ability to promote tumor cell proliferation, which appeared to be mediated via their production of neutrophil elastase, was rendered less effective. Relieving tumor hypoxia thus greatly improved net PMN-dependent tumor control, leading to a massive reduction in tumor burden. Remarkably, this outcome was T cell independent. Together, these findings identify key hypoxia-regulated molecular mechanisms through which PMNs directly induce tumor cell death and proliferation in vivo and suggest that the contrasting properties of PMNs in different tumor settings may in part reflect the effects of hypoxia on direct PMN-tumor cell interactions.
Collapse
Affiliation(s)
- Karim Mahiddine
- Department of Laboratory Medicine, UCSF, San Francisco, California, USA
| | - Adam Blaisdell
- Department of Pathology, NYU School of Medicine, New York, New York, USA
| | - Stephany Ma
- Department of Laboratory Medicine, UCSF, San Francisco, California, USA
| | | | - Clifford A Lowell
- Department of Laboratory Medicine, UCSF, San Francisco, California, USA.,Biomedical Sciences Program.,ImmunoX Program, and
| | - Adrian Erlebacher
- Department of Laboratory Medicine, UCSF, San Francisco, California, USA.,Biomedical Sciences Program.,ImmunoX Program, and.,Center for Reproductive Sciences, UCSF, San Francisco, California, USA
| |
Collapse
|
35
|
Liu Y, Wang J, Chen D, Kam WR, Sullivan DA. The Role of Hypoxia-Inducible Factor 1α in the Regulation of Human Meibomian Gland Epithelial Cells. Invest Ophthalmol Vis Sci 2020; 61:1. [PMID: 32150252 PMCID: PMC7401459 DOI: 10.1167/iovs.61.3.1] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022] Open
Abstract
Purpose We recently discovered that a hypoxic environment is beneficial for meibomian gland (MG) function. The mechanisms underlying this effect are unknown, but we hypothesize that it is due to an increase in the levels of hypoxia-inducible factor 1α (HIF1α). In other tissues, HIF1α is the primary regulator of cellular responses to hypoxia, and HIF1α expression can be induced by multiple stimuli, including hypoxia and hypoxia-mimetic agents. The objective of this study was to test our hypothesis. Methods Human eyelid tissues were stained for HIF1α. Immortalized human MG epithelial cells (IHMGECs) were cultured for varying time periods under normoxic (21% O2) or hypoxic (1% O2) conditions, in the presence or absence of the hypoxia-mimetic agent roxadustat (Roxa). IHMGECs were then processed for the analysis of cell number, HIF1α expression, lipid-containing vesicles, neutral and polar lipid content, DNase II activity, and intracellular pH. Results Our results show that HIF1α protein is present in human MG acinar epithelial cells in vivo. Our findings also demonstrate that exposure to 1% O2 or to Roxa increases the expression of HIF1α, the number of lipid-containing vesicles, the content of neutral lipids, and the activity of DNase II and decreases the pH in IHMGECs in vitro. Conclusions Our data support our hypothesis that the beneficial effect of hypoxia on the MG is mediated through an increased expression of HIF1α.
Collapse
|
36
|
de Almeida PE, Mak J, Hernandez G, Jesudason R, Herault A, Javinal V, Borneo J, Kim JM, Walsh KB. Anti-VEGF Treatment Enhances CD8 + T-cell Antitumor Activity by Amplifying Hypoxia. Cancer Immunol Res 2020; 8:806-818. [PMID: 32238381 DOI: 10.1158/2326-6066.cir-19-0360] [Citation(s) in RCA: 55] [Impact Index Per Article: 13.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2019] [Revised: 11/27/2019] [Accepted: 03/27/2020] [Indexed: 11/16/2022]
Abstract
Antiangiogenic therapies that target the VEGF pathway have been used clinically to combat cancer for over a decade. Beyond having a direct impact on blood vessel development and tumor perfusion, accumulating evidence indicates that these agents also affect antitumor immune responses. Numerous clinical trials combining antiangiogenic drugs with immunotherapies for the treatment of cancer are ongoing, but a mechanistic understanding of how disruption of tumor angiogenesis may impact immunity is not fully discerned. Here, we reveal that blockade of VEGF-A with a mAb to VEGF augments activation of CD8+ T cells within tumors and potentiates their capacity to produce cytokines. We demonstrate that this phenomenon relies on the disruption of VEGFR2 signaling in the tumor microenvironment but does not affect CD8+ T cells directly. Instead, the augmented functional capacity of CD8+ T cells stems from increased tumor hypoxia that initiates a hypoxia-inducible factor-1α program within CD8+ T cells that directly enhances cytokine production. Finally, combinatorial administration of anti-VEGF with an immunotherapeutic antibody, anti-OX40, improved antitumor activity over single-agent treatments. Our findings illustrate that anti-VEGF treatment enhances CD8+ T-cell effector function and provides a mechanistic rationale for combining antiangiogenic and immunotherapeutic drugs for cancer treatment.
Collapse
Affiliation(s)
| | - Judy Mak
- Department of Molecular Oncology, Genentech, Inc., South San Francisco, California
| | - Genevive Hernandez
- Oncology Biomarker Development, Genentech, Inc., South San Francisco, California
| | - Rajiv Jesudason
- Department of Molecular Oncology, Genentech, Inc., South San Francisco, California
| | - Aurelie Herault
- Department of Molecular Oncology, Genentech, Inc., South San Francisco, California
| | - Vincent Javinal
- Department of In-vivo Pharmacology, Genentech, Inc., South San Francisco, California
| | - Jovencio Borneo
- Department of Immunology and Infectious Diseases, Genentech, Inc., South San Francisco, California
| | - Jeong M Kim
- Department of Cancer Immunology, Genentech, Inc., South San Francisco, California
| | - Kevin B Walsh
- Department of Molecular Oncology, Genentech, Inc., South San Francisco, California.
| |
Collapse
|
37
|
Sharma M, Boytard L, Hadi T, Koelwyn G, Simon R, Ouimet M, Seifert L, Spiro W, Yan B, Hutchison S, Fisher EA, Ramasamy R, Ramkhelawon B, Moore KJ. Enhanced glycolysis and HIF-1α activation in adipose tissue macrophages sustains local and systemic interleukin-1β production in obesity. Sci Rep 2020; 10:5555. [PMID: 32221369 PMCID: PMC7101445 DOI: 10.1038/s41598-020-62272-9] [Citation(s) in RCA: 48] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2018] [Accepted: 03/10/2020] [Indexed: 12/23/2022] Open
Abstract
During obesity, macrophages infiltrate the visceral adipose tissue and promote inflammation that contributes to type II diabetes. Evidence suggests that the rewiring of cellular metabolism can regulate macrophage function. However, the metabolic programs that characterize adipose tissue macrophages (ATM) in obesity are poorly defined. Here, we demonstrate that ATM from obese mice exhibit metabolic profiles characterized by elevated glycolysis and oxidative phosphorylation, distinct from ATM from lean mice. Increased activation of HIF-1α in ATM of obese visceral adipose tissue resulted in induction of IL-1β and genes in the glycolytic pathway. Using a hypoxia-tracer, we show that HIF-1α nuclear translocation occurred both in hypoxic and non-hypoxic ATM suggesting that both hypoxic and pseudohypoxic stimuli activate HIF-1α and its target genes in ATM during diet-induced obesity. Exposure of macrophages to the saturated fatty acid palmitate increased glycolysis and HIF-1α expression, which culminated in IL-1β induction thereby simulating pseudohypoxia. Using mice with macrophage-specific targeted deletion of HIF-1α, we demonstrate the critical role of HIF-1α-derived from macrophages in regulating ATM accumulation, and local and systemic IL-1β production, but not in modulating systemic metabolic responses. Collectively, our data identify enhanced glycolysis and HIF-1α activation as drivers of low-grade inflammation in obesity.
Collapse
Affiliation(s)
- Monika Sharma
- Department of Medicine, Marc and Ruti Bell Program for Vascular Biology and Disease, The Leon H. Charney Division of Cardiology, New York University Langone Health, New York, NY, USA
| | - Ludovic Boytard
- Department of Surgery, New York University Langone Health, New York, NY, USA
| | - Tarik Hadi
- Department of Surgery, New York University Langone Health, New York, NY, USA
| | - Graeme Koelwyn
- Department of Medicine, Marc and Ruti Bell Program for Vascular Biology and Disease, The Leon H. Charney Division of Cardiology, New York University Langone Health, New York, NY, USA
| | - Russell Simon
- Department of Medicine, Marc and Ruti Bell Program for Vascular Biology and Disease, The Leon H. Charney Division of Cardiology, New York University Langone Health, New York, NY, USA
| | - Mireille Ouimet
- Department of Medicine, Marc and Ruti Bell Program for Vascular Biology and Disease, The Leon H. Charney Division of Cardiology, New York University Langone Health, New York, NY, USA
| | - Lena Seifert
- Department of Surgery, New York University Langone Health, New York, NY, USA
| | - Westley Spiro
- Department of Medicine, Marc and Ruti Bell Program for Vascular Biology and Disease, The Leon H. Charney Division of Cardiology, New York University Langone Health, New York, NY, USA
| | - Bo Yan
- Department of Medicine, Marc and Ruti Bell Program for Vascular Biology and Disease, The Leon H. Charney Division of Cardiology, New York University Langone Health, New York, NY, USA
| | - Susan Hutchison
- Department of Medicine, Marc and Ruti Bell Program for Vascular Biology and Disease, The Leon H. Charney Division of Cardiology, New York University Langone Health, New York, NY, USA
| | - Edward A Fisher
- Department of Medicine, Marc and Ruti Bell Program for Vascular Biology and Disease, The Leon H. Charney Division of Cardiology, New York University Langone Health, New York, NY, USA
| | - Ravichandran Ramasamy
- Department of Medicine, Division of Endocrinology, New York University Langone Health, New York, NY, USA
| | - Bhama Ramkhelawon
- Department of Surgery, New York University Langone Health, New York, NY, USA.
| | - Kathryn J Moore
- Department of Medicine, Marc and Ruti Bell Program for Vascular Biology and Disease, The Leon H. Charney Division of Cardiology, New York University Langone Health, New York, NY, USA.
| |
Collapse
|
38
|
Macklin PS, Yamamoto A, Browning L, Hofer M, Adam J, Pugh CW. Recent advances in the biology of tumour hypoxia with relevance to diagnostic practice and tissue-based research. J Pathol 2020; 250:593-611. [PMID: 32086807 DOI: 10.1002/path.5402] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2020] [Accepted: 02/18/2020] [Indexed: 02/06/2023]
Abstract
In this review article, we examine the importance of low levels of oxygen (hypoxia) in cancer biology. We provide a brief description of how mammalian cells sense oxygen. The hypoxia-inducible factor (HIF) pathway is currently the best characterised oxygen-sensing system, but recent work has revealed that mammals also use an oxygen-sensing system found in plants to regulate the abundance of some proteins and peptides with an amino-terminal cysteine residue. We discuss how the HIF pathway is affected during the growth of solid tumours, which develop in microenvironments with gradients of oxygen availability. We then introduce the concept of 'pseudohypoxia', a state of constitutive, oxygen-independent HIF system activation that occurs due to oncogenic stimulation in a number of specific tumour types that are of immediate relevance to diagnostic histopathologists. We provide an overview of the different methods of quantifying tumour hypoxia, emphasising the importance of pre-analytic factors in interpreting the results of tissue-based studies. Finally, we review recent approaches to targeting hypoxia/HIF system activation for therapeutic benefit, the application of which may require knowledge of which hypoxia signalling components are being utilised by a given tumour. © 2020 The Authors. The Journal of Pathology published by John Wiley & Sons Ltd on behalf of Pathological Society of Great Britain and Ireland.
Collapse
Affiliation(s)
- Philip S Macklin
- Nuffield Department of Medicine, University of Oxford, Oxford, UK
| | - Atsushi Yamamoto
- Nuffield Department of Medicine, University of Oxford, Oxford, UK
| | - Lisa Browning
- Department of Cellular Pathology, Oxford University Hospitals NHS Foundation Trust, Oxford, UK
| | - Monika Hofer
- Department of Neuropathology and Ocular Pathology, Oxford University Hospitals NHS Foundation Trust, Oxford, UK
| | - Julie Adam
- Nuffield Department of Medicine, University of Oxford, Oxford, UK
| | | |
Collapse
|
39
|
Buchholz B, Eckardt KU. Role of oxygen and the HIF-pathway in polycystic kidney disease. Cell Signal 2020; 69:109524. [PMID: 31904413 DOI: 10.1016/j.cellsig.2020.109524] [Citation(s) in RCA: 24] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2019] [Revised: 01/01/2020] [Accepted: 01/01/2020] [Indexed: 12/16/2022]
Abstract
Kidney cyst growth in ADPKD is associated with regional hypoxia, presumably due to a mismatch between enlarged cysts and the peritubular capillary blood supply and compression of peritubular capillaries in cyst walls. Regional hypoxia leads to activation of hypoxia-inducible transcription factors, with the two main HIF isoforms, HIF-1 and HIF-2 expressed in cyst epithelia and pericystic interstitial cells, respectively. While HIF-2 activation is linked to EPO production, mitigating the anemia that normally accompanies chronic kidney disease, HIF-1 promotes cyst growth. HIF-dependent cyst growth is primarily due to an increase in chloride-dependent fluid secretion into the cyst lumen. However, given the broad spectrum of HIF-target genes, additional HIF-mediated pathways may also contribute to cyst progression. Furthermore, hypoxia can influence cyst growth through the generation of reactive oxygen species. Since cyst expansion aggravates regional hypoxia, a feedforward loop is established that accelerates cyst expansion and disease progression. Inhibiting the HIF pathway and/or HIF target genes that are of particular relevance for HIF-dependent cyst fluid secretion may therefore represent novel therapeutic approaches to retard the progression of APDKD.
Collapse
Affiliation(s)
- Bjoern Buchholz
- Department of Nephrology and Hypertension, Friedrich-Alexander-Universität Erlangen-Nürnberg, Erlangen, Germany.
| | - Kai-Uwe Eckardt
- Department of Nephrology and Medical Intensive Care, Charité, Universitätsmedizin Berlin, Berlin, Germany
| |
Collapse
|
40
|
Kim R, Attayek PJ, Wang Y, Furtado KL, Tamayo R, Sims CE, Allbritton NL. An in vitro intestinal platform with a self-sustaining oxygen gradient to study the human gut/microbiome interface. Biofabrication 2019; 12:015006. [PMID: 31519008 PMCID: PMC6933551 DOI: 10.1088/1758-5090/ab446e] [Citation(s) in RCA: 64] [Impact Index Per Article: 12.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
An oxygen gradient formed along the length of colonic crypts supports stem-cell proliferation at the normoxic crypt base while supporting obligate anaerobe growth in the anoxic colonic lumen. Primary human colonic epithelial cells derived from human gastrointestinal stem cells were cultured within a device possessing materials of tailored oxygen permeability to produce an oxygen-depleted luminal (0.8% ± 0.1% O2) and oxygen-rich basal (11.1% ± 0.5% O2) compartment. This oxygen difference created a stable oxygen gradient across the colonic epithelial cells which remained viable and properly polarized. Facultative and obligate anaerobes Lactobacillus rhamnosus, Bifidobacterium adolescentis, and Clostridium difficile grew readily within the luminal compartment. When formed along the length of an in vitro crypt, the oxygen gradient facilitated cell compartmentalization within the crypt by enhancing confinement of the proliferative cells to the crypt base. This platform provides a simple system to create a physiological oxygen gradient across an intestinal mimic while simultaneously supporting anaerobe co-culture.
Collapse
Affiliation(s)
- Raehyun Kim
- Joint Department of Biomedical Engineering, University of North Carolina, Chapel Hill, and North Carolina State University, Raleigh, North Carolina
| | - Peter J. Attayek
- Joint Department of Biomedical Engineering, University of North Carolina, Chapel Hill, and North Carolina State University, Raleigh, North Carolina
| | - Yuli Wang
- Department of Chemistry, University of North Carolina, Chapel Hill, North Carolina
| | - Kathleen L. Furtado
- Department of Microbiology and Immunology, University of North Carolina, Chapel Hill, North Carolina
| | - Rita Tamayo
- Department of Microbiology and Immunology, University of North Carolina, Chapel Hill, North Carolina
| | - Christopher E. Sims
- Department of Chemistry, University of North Carolina, Chapel Hill, North Carolina
| | - Nancy L. Allbritton
- Joint Department of Biomedical Engineering, University of North Carolina, Chapel Hill, and North Carolina State University, Raleigh, North Carolina
- Department of Chemistry, University of North Carolina, Chapel Hill, North Carolina
| |
Collapse
|
41
|
Ow CPC, Ullah MM, Ngo JP, Sayakkarage A, Evans RG. Detection of cellular hypoxia by pimonidazole adduct immunohistochemistry in kidney disease: methodological pitfalls and their solution. Am J Physiol Renal Physiol 2019; 317:F322-F332. [PMID: 31188031 DOI: 10.1152/ajprenal.00219.2019] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023] Open
Abstract
Pimonidazole adduct immunohistochemistry is one of the few available methods for assessing renal tissue hypoxia at the cellular level. It appears to be prone to artifactual false positive staining under some circumstances. Here, we assessed the nature of this false positive staining and, having determined how to avoid it, reexamined the nature of cellular hypoxia in rat models of kidney disease. When a mouse-derived anti-pimonidazole primary antibody was used, two types of staining were observed. First, there was diffuse staining of the cytoplasm of tubular epithelial cells, which was largely absent when the primary antibody was omitted from the incubation protocol or in tissues known not to contain pimonidazole adducts. Second, there was staining of the apical membranes of tubular epithelial cells, debris within the lumen of renal tubules, including tubular casts, and the interstitium; this latter staining was present even when the primary antibody was omitted from the incubation protocol. Such false positive staining was particularly prominent in acutely injured kidneys. It could not be avoided by preincubation of sections with a mouse IgG blocking reagent. Furthermore, preadsorption of the secondary antibody against rat Ig abolished all staining; however, when a rabbit-derived polyclonal anti-pimonidazole primary antibody was used, the false positive staining was largely avoided. Using this method, we confirmed the presence of hypoxia, localized mainly to the tubular epithelium, in the acute phase of severe renal ischemia-reperfusion injury, adenine-induced chronic kidney disease, and polycystic kidney disease. We conclude that this new method provides improved detection of renal cellular hypoxia.
Collapse
Affiliation(s)
- Connie P C Ow
- Cardiovascular Disease Program, Biomedicine Discovery Institute and Department of Physiology, Monash University , Melbourne, Victoria , Australia
| | - Md Mahbub Ullah
- Cardiovascular Disease Program, Biomedicine Discovery Institute and Department of Physiology, Monash University , Melbourne, Victoria , Australia
| | - Jennifer P Ngo
- Cardiovascular Disease Program, Biomedicine Discovery Institute and Department of Physiology, Monash University , Melbourne, Victoria , Australia
| | - Adheeshee Sayakkarage
- Cardiovascular Disease Program, Biomedicine Discovery Institute and Department of Physiology, Monash University , Melbourne, Victoria , Australia
| | - Roger G Evans
- Cardiovascular Disease Program, Biomedicine Discovery Institute and Department of Physiology, Monash University , Melbourne, Victoria , Australia
| |
Collapse
|
42
|
A platinum(II)-acetylide-based conjugated polyelectrolyte for hypoxia imaging via ratiometric and time-resolved luminescence microscopy. J Organomet Chem 2019. [DOI: 10.1016/j.jorganchem.2018.10.014] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/01/2023]
|
43
|
Liu Y, Chen D, Chen X, Kam WR, Hatton MP, Sullivan DA. Hypoxia: A breath of fresh air for the meibomian gland. Ocul Surf 2018; 17:310-317. [PMID: 30528291 DOI: 10.1016/j.jtos.2018.12.001] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2018] [Revised: 11/30/2018] [Accepted: 12/03/2018] [Indexed: 12/19/2022]
Abstract
PURPOSE Optimal meibomian gland (MG) function is critically important for the health and wellbeing of the ocular surface. We hypothesize that low oxygen (O2) conditions promote the function of human MG epithelial cells (HMGECs) and that human MGs exist in a relatively hypoxic environment. The purpose of this study was to test our hypotheses. METHODS We used human and mouse eyelid segments, and immortalized human MG epithelial cells (IHMGECs) in our studies. To evaluate oxygen (O2) levels in the mouse MG and vicinity, we injected pimonidazole (pimo), a hypoxia marker, before sacrifice. Human eyelid samples were stained with the hypoxia marker glucose transporter 1 (Glut-1). To determine the effect of low O2 levels on IHMGECs, we cultured cells under proliferating and differentiating conditions in both normoxic (21% O2) and hypoxic (3% O2) conditions for 5-15 days. IHMGECs were evaluated for cell number, neutral lipid content, lysosome accumulation, expression of biomarker proteins and DNase II activity. RESULTS Our results demonstrate that human and mouse MGs, but not the surrounding connective tissue, exist in a relatively hypoxic environment in vivo. In addition, our findings show that hypoxia does not influence IHMGEC numbers in basal or proliferating culture conditions, but does stimulate the expression of SREBP-1 in differentiating IHMGECs. Hypoxia also significantly increased DNase II activity, and apparently IHMGEC terminal differentiation. CONCLUSIONS Our Results support our hypotheses, and indicate that relative hypoxia promotes MG function.
Collapse
Affiliation(s)
- Yang Liu
- Schepens Eye Research Institute, Massachusetts Eye and Ear, and Department of Ophthalmology, Harvard Medical School, Boston, 02114, USA.
| | - Di Chen
- Schepens Eye Research Institute, Massachusetts Eye and Ear, and Department of Ophthalmology, Harvard Medical School, Boston, 02114, USA; Department of Ophthalmology, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, 100730, China
| | - Xiaomin Chen
- Schepens Eye Research Institute, Massachusetts Eye and Ear, and Department of Ophthalmology, Harvard Medical School, Boston, 02114, USA; Zhongnan Hospital, Wuhan University, Wuhan, China
| | - Wendy R Kam
- Schepens Eye Research Institute, Massachusetts Eye and Ear, and Department of Ophthalmology, Harvard Medical School, Boston, 02114, USA
| | - Mark P Hatton
- Schepens Eye Research Institute, Massachusetts Eye and Ear, and Department of Ophthalmology, Harvard Medical School, Boston, 02114, USA; Ophthalmic Consultants of Boston, Boston, 02114, USA
| | - David A Sullivan
- Schepens Eye Research Institute, Massachusetts Eye and Ear, and Department of Ophthalmology, Harvard Medical School, Boston, 02114, USA
| |
Collapse
|
44
|
Moosvi F, Baker JH, Yung A, Kozlowski P, Minchinton AI, Reinsberg SA. Fast and sensitive dynamic oxygen‐enhanced MRI with a cycling gas challenge and independent component analysis. Magn Reson Med 2018; 81:2514-2525. [DOI: 10.1002/mrm.27584] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2018] [Revised: 10/04/2018] [Accepted: 10/07/2018] [Indexed: 12/14/2022]
Affiliation(s)
- Firas Moosvi
- Department of Physics & Astronomy University of British Columbia Vancouver Canada
| | - Jennifer H.E. Baker
- Radiation Biology Unit British Columbia Cancer Research Centre Vancouver Canada
| | - Andrew Yung
- UBC MRI Research Centre Life Sciences Centre Vancouver Canada
| | - Piotr Kozlowski
- UBC MRI Research Centre Life Sciences Centre Vancouver Canada
- Department of Radiology University of British Columbia Vancouver Canada
| | | | - Stefan A. Reinsberg
- Department of Physics & Astronomy University of British Columbia Vancouver Canada
| |
Collapse
|
45
|
Marsboom G, Rehman J. Hypoxia Signaling in Vascular Homeostasis. Physiology (Bethesda) 2018; 33:328-337. [PMID: 30109825 PMCID: PMC6230550 DOI: 10.1152/physiol.00018.2018] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2018] [Revised: 07/09/2018] [Accepted: 07/09/2018] [Indexed: 01/10/2023] Open
Abstract
Hypoxia signaling in the vasculature controls vascular permeability, inflammation, vascular growth, and repair of vascular injury. In this review, we summarize recent insights in this burgeoning field and highlight the importance of studying the heterogeneity of hypoxia responses among individual patients, distinct vascular beds, and even individual vascular cells.
Collapse
Affiliation(s)
- Glenn Marsboom
- Department of Pharmacology, University of Illinois College of Medicine , Chicago, Illinois
| | - Jalees Rehman
- Department of Pharmacology, University of Illinois College of Medicine , Chicago, Illinois
- Department of Medicine, Section of Cardiology, University of Illinois College of Medicine , Chicago, Illinois
| |
Collapse
|
46
|
Dheeraj A, Agarwal C, Schlaepfer IR, Raben D, Singh R, Agarwal R, Deep G. A novel approach to target hypoxic cancer cells via combining β-oxidation inhibitor etomoxir with radiation. HYPOXIA 2018; 6:23-33. [PMID: 30175155 PMCID: PMC6109663 DOI: 10.2147/hp.s163115] [Citation(s) in RCA: 34] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 01/08/2023]
Abstract
Background Hypoxia in tumors is associated with resistance towards various therapies including radiotherapy. In this study, we assessed if hypoxia in cancer spheres could be effectively reduced by adding etomoxir (a β-oxidation inhibitor) immediately after cell irradiation. Methods We employed cancer cells’ sphere model to target hypoxia. Confocal imaging was used to analyze hypoxia and expression of specific biomarkers in spheres following various treatments (radiation and/or etomoxir). Results Etomoxir (32.5 μM) treatment improved the radiation (2.5 Gy) efficacy against growth of lung adenocarcinoma H460 spheres. More importantly, radiation and etomoxir combination significantly reduced the hypoxic regions (pimonidazole+ areas) in H460 spheres compared to either treatment alone. Also, etomoxir and radiation combination treatment reduced the protein level of biomarkers for proliferation (Ki-67 and cyclin D1), stemness (CD44) and β-oxidation (CPT1A) in H460 spheres. We observed similar efficacy of etomoxir against growth of prostate cancer LNCaP cells’ spheres when combined with radiation. Further, radiation treatment strongly reduced the hypoxic regions (pimonidazole+ areas) in CPT1 knockdown LNCaP cells’ spheres. Conclusions Together, these results offer a unique approach to target hypoxia in solid tumors via combining etomoxir with radiation, thereby improving therapeutic efficacy.
Collapse
Affiliation(s)
- Arpit Dheeraj
- Department of Pharmaceutical Sciences, Skaggs School of Pharmacy and Pharmaceutical Sciences, University of Colorado Anschutz Medical Campus, Aurora, CO, USA.,School of Life Sciences, Jawaharlal Nehru University, New Delhi, India
| | - Chapla Agarwal
- Department of Pharmaceutical Sciences, Skaggs School of Pharmacy and Pharmaceutical Sciences, University of Colorado Anschutz Medical Campus, Aurora, CO, USA
| | - Isabel R Schlaepfer
- Division of Medical Oncology, University of Colorado Anschutz Medical Campus, Aurora, CO, USA
| | - David Raben
- Department of Radiation Oncology, University of Colorado Anschutz Medical Campus, Aurora, CO, USA
| | - Rana Singh
- School of Life Sciences, Jawaharlal Nehru University, New Delhi, India
| | - Rajesh Agarwal
- Department of Pharmaceutical Sciences, Skaggs School of Pharmacy and Pharmaceutical Sciences, University of Colorado Anschutz Medical Campus, Aurora, CO, USA
| | - Gagan Deep
- Department of Cancer Biology, Wake Forest School of Medicine, Winston-Salem, NC, USA, .,Department of Urology, Wake Forest School of Medicine, Winston-Salem, NC, USA, .,Wake Forest Baptist Comprehensive Cancer Center, Winston-Salem, NC, USA,
| |
Collapse
|
47
|
Yasui H, Kawai T, Matsumoto S, Saito K, Devasahayam N, Mitchell JB, Camphausen K, Inanami O, Krishna MC. Quantitative imaging of pO 2 in orthotopic murine gliomas: hypoxia correlates with resistance to radiation. Free Radic Res 2018; 51:861-871. [PMID: 29076398 DOI: 10.1080/10715762.2017.1388506] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022]
Abstract
Hypoxia is considered one of the microenvironmental factors associated with the malignant nature of glioblastoma. Thus, evaluating intratumoural distribution of hypoxia would be useful for therapeutic planning as well as assessment of its effectiveness during the therapy. Electron paramagnetic resonance imaging (EPRI) is an imaging technique which can generate quantitative maps of oxygen in vivo using the exogenous paramagnetic compound, triarylmethyl and monitoring its line broadening caused by oxygen. In this study, the feasibility of EPRI for assessment of oxygen distribution in the glioblastoma using orthotopic U87 and U251 xenograft model is examined. Heterogeneous distribution of pO2 between 0 and 50 mmHg was observed throughout the tumours except for the normal brain tissue. U251 glioblastoma was more likely to exhibit hypoxia than U87 for comparable tumour size (median pO2; 29.7 and 18.2 mmHg, p = .028, in U87 and U251, respectively). The area with pO2 under 10 mmHg on the EPR oximetry (HF10) showed a good correlation with pimonidazole staining among tumours with evaluated size. In subcutaneous xenograft model, irradiation was relatively less effective for U251 compared with U87. In conclusion, EPRI is a feasible method to evaluate oxygen distribution in the brain tumour.
Collapse
Affiliation(s)
- Hironobu Yasui
- a Central Institute of Isotope Science, Hokkaido University , Sapporo , Japan
| | - Tatsuya Kawai
- b Radiation Oncology Branch , Center for Cancer Research, National Cancer Institute, National Health Institutes , Bethesda , MD , USA
| | - Shingo Matsumoto
- c Division of Bioengineering and Bioinformatics , Graduate School of Information Science and Technology, Hokkaido University , Sapporo , Japan
| | - Keita Saito
- d Radiation Biology Branch , Center for Cancer Research, National Cancer Institute, National Health Institutes , Bethesda , MD , USA
| | - Nallathamby Devasahayam
- d Radiation Biology Branch , Center for Cancer Research, National Cancer Institute, National Health Institutes , Bethesda , MD , USA
| | - James B Mitchell
- d Radiation Biology Branch , Center for Cancer Research, National Cancer Institute, National Health Institutes , Bethesda , MD , USA
| | - Kevin Camphausen
- b Radiation Oncology Branch , Center for Cancer Research, National Cancer Institute, National Health Institutes , Bethesda , MD , USA
| | - Osamu Inanami
- e Laboratory of Radiation Biology, Department of Environmental Veterinary Sciences, Graduate School of Veterinary Medicine , Hokkaido University , Sapporo , Japan
| | - Murali C Krishna
- d Radiation Biology Branch , Center for Cancer Research, National Cancer Institute, National Health Institutes , Bethesda , MD , USA
| |
Collapse
|
48
|
Read GH, Miura N, Carter JL, Kines KT, Yamamoto K, Devasahayam N, Cheng JY, Camphausen KA, Krishna MC, Kesarwala AH. Three-dimensional alginate hydrogels for radiobiological and metabolic studies of cancer cells. Colloids Surf B Biointerfaces 2018; 171:197-204. [PMID: 30031304 DOI: 10.1016/j.colsurfb.2018.06.018] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2017] [Revised: 04/23/2018] [Accepted: 06/14/2018] [Indexed: 01/05/2023]
Abstract
The purpose of this study is to demonstrate calcium alginate hydrogels as a system for in vitro radiobiological and metabolic studies of cancer cells. Previous studies have established calcium alginate as a versatile three-dimensional (3D) culturing system capable of generating areas of oxygen heterogeneity and modeling metabolic changes in vitro. Here, through dosimetry, clonogenic and viability assays, and pimonidazole staining, we demonstrate that alginate can model radiobiological responses that monolayer cultures do not simulate. Notably, alginate hydrogels with radii greater than 500 μm demonstrate hypoxic cores, while smaller hydrogels do not. The size of this hypoxic region correlates with hydrogel size and improved cell survival following radiation therapy. Hydrogels can also be utilized in hyperpolarized magnetic resonance spectroscopy and extracellular flux analysis. Alginate therefore offers a reproducible, consistent, and low-cost means for 3D culture of cancer cells for radiobiological studies that simulates important in vivo parameters such as regional hypoxia and enables long-term culturing and in vitro metabolic studies.
Collapse
Affiliation(s)
- Graham H Read
- Radiation Oncology Branch, National Cancer Institute, National Institutes of Health, Bethesda, MD, 20892, USA
| | - Natsuko Miura
- Radiation Oncology Branch, National Cancer Institute, National Institutes of Health, Bethesda, MD, 20892, USA
| | - Jenna L Carter
- Radiation Oncology Branch, National Cancer Institute, National Institutes of Health, Bethesda, MD, 20892, USA
| | - Kelsey T Kines
- Radiation Oncology Branch, National Cancer Institute, National Institutes of Health, Bethesda, MD, 20892, USA
| | - Kazutoshi Yamamoto
- Radiation Biology Branch, National Cancer Institute, National Institutes of Health, Bethesda, MD, 20892, USA
| | - Nallathamby Devasahayam
- Radiation Biology Branch, National Cancer Institute, National Institutes of Health, Bethesda, MD, 20892, USA
| | - Jason Y Cheng
- Radiation Oncology Branch, National Cancer Institute, National Institutes of Health, Bethesda, MD, 20892, USA
| | - Kevin A Camphausen
- Radiation Oncology Branch, National Cancer Institute, National Institutes of Health, Bethesda, MD, 20892, USA
| | - Murali C Krishna
- Radiation Biology Branch, National Cancer Institute, National Institutes of Health, Bethesda, MD, 20892, USA
| | - Aparna H Kesarwala
- Radiation Oncology Branch, National Cancer Institute, National Institutes of Health, Bethesda, MD, 20892, USA; Lead Contact, USA.
| |
Collapse
|
49
|
Borton AH, Benson BL, Neilson LE, Saunders A, Alaiti MA, Huang AY, Jain MK, Proweller A, Ramirez-Bergeron DL. Aryl Hydrocarbon Receptor Nuclear Translocator in Vascular Smooth Muscle Cells Is Required for Optimal Peripheral Perfusion Recovery. J Am Heart Assoc 2018; 7:e009205. [PMID: 29858371 PMCID: PMC6015385 DOI: 10.1161/jaha.118.009205] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/20/2018] [Accepted: 05/02/2018] [Indexed: 11/23/2022]
Abstract
BACKGROUND Limb ischemia resulting from peripheral vascular disease is a common cause of morbidity. Vessel occlusion limits blood flow, creating a hypoxic environment that damages distal tissue, requiring therapeutic revascularization. Hypoxia-inducible factors (HIFs) are key transcriptional regulators of hypoxic vascular responses, including angiogenesis and arteriogenesis. Despite vascular smooth muscle cells' (VSMCs') importance in vessel integrity, little is known about their functional responses to hypoxia in peripheral vascular disease. This study investigated the role of VSMC HIF in mediating peripheral ischemic responses. METHODS AND RESULTS We used ArntSMKO mice with smooth muscle-specific deletion of aryl hydrocarbon receptor nuclear translocator (ARNT, HIF-1β), required for HIF transcriptional activity, in a femoral artery ligation model of peripheral vascular disease. ArntSMKO mice exhibit impaired perfusion recovery despite normal collateral vessel dilation and angiogenic capillary responses. Decreased blood flow manifests in extensive tissue damage and hypoxia in ligated limbs of ArntSMKO mice. Furthermore, loss of aryl hydrocarbon receptor nuclear translocator changes the proliferation, migration, and transcriptional profile of cultured VSMCs. ArntSMKO mice display disrupted VSMC morphologic features and wrapping around arterioles and increased vascular permeability linked to decreased local blood flow. CONCLUSIONS Our data demonstrate that traditional vascular remodeling responses are insufficient to provide robust peripheral tissue reperfusion in ArntSMKO mice. In all, this study highlights HIF responses to hypoxia in arteriole VSMCs critical for the phenotypic and functional stability of vessels that aid in the recovery of blood flow in ischemic peripheral tissues.
Collapse
MESH Headings
- Animals
- Aryl Hydrocarbon Receptor Nuclear Translocator/biosynthesis
- Aryl Hydrocarbon Receptor Nuclear Translocator/genetics
- Blotting, Western
- Cells, Cultured
- Disease Models, Animal
- Gene Expression Regulation
- Immunohistochemistry
- Ischemia/genetics
- Ischemia/metabolism
- Ischemia/pathology
- Lower Extremity/blood supply
- Mice
- Mice, Inbred C57BL
- Mice, Knockout
- Microscopy, Confocal
- Muscle, Smooth, Vascular/metabolism
- Muscle, Smooth, Vascular/pathology
- Peripheral Vascular Diseases/genetics
- Peripheral Vascular Diseases/metabolism
- Peripheral Vascular Diseases/pathology
- RNA/genetics
- Reverse Transcriptase Polymerase Chain Reaction
Collapse
Affiliation(s)
- Anna Henry Borton
- Department of Pathology, Case Western Reserve University School of Medicine, Cleveland, OH
- Case Cardiovascular Research Institute, Case Western Reserve University School of Medicine, Cleveland, OH
- Harrington Heart and Vascular Institute, University Hospitals, Cleveland, OH
| | - Bryan L Benson
- Department of Pathology, Case Western Reserve University School of Medicine, Cleveland, OH
| | - Lee E Neilson
- Neurological Institute, University Hospitals, Cleveland, OH
| | - Ashley Saunders
- Case Cardiovascular Research Institute, Case Western Reserve University School of Medicine, Cleveland, OH
- Harrington Heart and Vascular Institute, University Hospitals, Cleveland, OH
| | - M Amer Alaiti
- Case Cardiovascular Research Institute, Case Western Reserve University School of Medicine, Cleveland, OH
- Harrington Heart and Vascular Institute, University Hospitals, Cleveland, OH
| | - Alex Y Huang
- Department of Pathology, Case Western Reserve University School of Medicine, Cleveland, OH
- Division of Pediatric Hematology-Oncology, Department of Pediatrics, Case Western Reserve University School of Medicine, Cleveland, OH
- Case Comprehensive Cancer Center, Case Western Reserve University School of Medicine, Cleveland, OH
- Angie Fowler Adolescent and Young Adult Cancer Institute and University Hospitals Rainbow Babies and Children's Hospital University Hospitals, Cleveland, OH
| | - Mukesh K Jain
- Case Cardiovascular Research Institute, Case Western Reserve University School of Medicine, Cleveland, OH
- Harrington Heart and Vascular Institute, University Hospitals, Cleveland, OH
| | - Aaron Proweller
- Case Cardiovascular Research Institute, Case Western Reserve University School of Medicine, Cleveland, OH
- Harrington Heart and Vascular Institute, University Hospitals, Cleveland, OH
| | - Diana L Ramirez-Bergeron
- Case Cardiovascular Research Institute, Case Western Reserve University School of Medicine, Cleveland, OH
- Harrington Heart and Vascular Institute, University Hospitals, Cleveland, OH
| |
Collapse
|
50
|
Kim H, Lin Q, Yun Z. The hypoxic tumor microenvironment in vivo selects tumor cells with increased survival against genotoxic stresses. Cancer Lett 2018; 431:142-149. [PMID: 29859297 DOI: 10.1016/j.canlet.2018.05.047] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2018] [Revised: 05/24/2018] [Accepted: 05/25/2018] [Indexed: 11/28/2022]
Abstract
Tumor sensitivity to radiation therapy has been known to be dependent on O2 concentrations. However, radiosensitivity of naturally occurring hypoxic tumor cells remains to be well fully investigated in direct comparison to that of their adjacent non-hypoxic tumor cells within the same tumor. We developed a hypoxia-sensing xenograft model using the hypoxia-response element (HRE)-driven enhanced green fluorescence protein (EGFP) as a hypoxia reporter to identify hypoxic tumor cells in situ. Here, we have found that naturally hypoxic tumor cells are moderately radioresistant compared to their neighboring non-hypoxic tumor cells in the same tumor. These naturally hypoxic tumor cells are proficient at repairing DNA damages and resist apoptosis induced by genotoxic stresses, which involves activation of the ATM/CHK1/CHK2 DNA damage-sensing pathway. Inhibition of the checkpoint kinases sensitizes the ex vivo hypoxic tumor cells to ionizing irradiation. Second, the new functional phenotypes acquired by the hypoxic tumor cells in vivo are stable even after they are maintained under non-hypoxic conditions. These new results strongly suggest that the hypoxic tumor microenvironment is capable of selecting stable tumor cell populations with increased resistance to genotoxic stresses and enhanced survival.
Collapse
Affiliation(s)
- Hoon Kim
- Department of Therapeutic Radiology, Yale School of Medicine, New Haven, CT, 06510, USA
| | - Qun Lin
- Department of Therapeutic Radiology, Yale School of Medicine, New Haven, CT, 06510, USA
| | - Zhong Yun
- Department of Therapeutic Radiology, Yale School of Medicine, New Haven, CT, 06510, USA.
| |
Collapse
|