1
|
Singh RK, Torne AS, Robertson ES. Hypoxic reactivation of Kaposi's sarcoma associated herpesvirus. CELL INSIGHT 2024; 3:100200. [PMID: 39391006 PMCID: PMC11466537 DOI: 10.1016/j.cellin.2024.100200] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/30/2024] [Revised: 08/28/2024] [Accepted: 08/29/2024] [Indexed: 10/12/2024]
Abstract
Hypoxic reactivation of Kaposi's sarcoma-associated herpesvirus (KSHV) refers to the phenomenon under low oxygen where the virus goes from latent to lytic replication. Typically, healthy cells generally cease cell division and DNA replication under hypoxic conditions due to limited resources, and the presence of physiological inhibitors. This restricted replication under hypoxic conditions is considered an employed strategy of the cell to minimize energy consumption. However, cancerous cells continuously replicate and divide in hypoxic conditions by reprogramming several aspects of their cell physiology, including but not limited to metabolism, cell cycle, DNA replication, transcription, translation, and the epigenome. KSHV infection, similar to cancerous cells, is known to bypass hypoxia-induced restrictions and undergo reactivation to produce progeny viruses. In previous studies we have mapped several aspects of cell physiology that are manipulated by KSHV through its latent antigens during hypoxic conditions, which allows for a permissive environment for its replication. We discuss the major strategies utilized by KSHV to bypass hypoxia-induced repression. We also describe the KSHV-encoded antigens responsible for modulating these cellular processes important for successful viral replication and persistence in hypoxia.
Collapse
Affiliation(s)
- Rajnish Kumar Singh
- Department of Otorhinolaryngology-Head and Neck Surgery, Perelman School of Medicine, University of Pennsylvania, Philadelphia, 19104, USA
| | - Atharva S Torne
- Department of Otorhinolaryngology-Head and Neck Surgery, Perelman School of Medicine, University of Pennsylvania, Philadelphia, 19104, USA
| | - Erle S Robertson
- Department of Otorhinolaryngology-Head and Neck Surgery, Perelman School of Medicine, University of Pennsylvania, Philadelphia, 19104, USA
| |
Collapse
|
2
|
Advani D, Kumar P. Uncovering Cell Cycle Dysregulations and Associated Mechanisms in Cancer and Neurodegenerative Disorders: A Glimpse of Hope for Repurposed Drugs. Mol Neurobiol 2024; 61:8600-8630. [PMID: 38532240 DOI: 10.1007/s12035-024-04130-7] [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: 12/25/2023] [Accepted: 03/19/2024] [Indexed: 03/28/2024]
Abstract
The cell cycle is the sequence of events orchestrated by a complex network of cell cycle proteins. Unlike normal cells, mature neurons subsist in a quiescent state of the cell cycle, and aberrant cell cycle activation triggers neuronal death accompanied by neurodegeneration. The periodicity of cell cycle events is choreographed by various mechanisms, including DNA damage repair, oxidative stress, neurotrophin activity, and ubiquitin-mediated degradation. Given the relevance of cell cycle processes in cancer and neurodegeneration, this review delineates the overlapping cell cycle events, signaling pathways, and mechanisms associated with cell cycle aberrations in cancer and the major neurodegenerative disorders. We suggest that dysregulation of some common fundamental signaling processes triggers anomalous cell cycle activation in cancer cells and neurons. We discussed the possible use of cell cycle inhibitors for neurodegenerative disorders and described the associated challenges. We propose that a greater understanding of the common mechanisms driving cell cycle aberrations in cancer and neurodegenerative disorders will open a new avenue for the development of repurposed drugs.
Collapse
Affiliation(s)
- Dia Advani
- Molecular Neuroscience and Functional Genomics Laboratory, Department of Biotechnology, Delhi Technological University (Formerly Delhi College of Engineering), Shahbad Daulatpur, Bawana Road, New Delhi, Delhi, 110042, India
| | - Pravir Kumar
- Molecular Neuroscience and Functional Genomics Laboratory, Department of Biotechnology, Delhi Technological University (Formerly Delhi College of Engineering), Shahbad Daulatpur, Bawana Road, New Delhi, Delhi, 110042, India.
| |
Collapse
|
3
|
Lee Y, Byeon E, Lee JS, Maszczyk P, Kim HS, Sayed AEDH, Yang Z, Lee JS, Kim DH. Differential susceptibility to hypoxia in hypoxia-inducible factor 1-alpha (HIF-1α)-targeted freshwater water flea Daphnia magna mutants. MARINE POLLUTION BULLETIN 2024; 209:117138. [PMID: 39486200 DOI: 10.1016/j.marpolbul.2024.117138] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/11/2024] [Revised: 10/10/2024] [Accepted: 10/10/2024] [Indexed: 11/04/2024]
Abstract
The water flea, Daphnia magna, serves as a key model organism for investigating the response of aquatic organisms to environmental stressors, including hypoxia. Hypoxia-inducible factor 1-alpha (HIF-1α) is a central regulatory protein involved in the cellular response to hypoxic conditions. In this study, we used CRISPR/Cas9 gene editing to create D. magna mutant lines with targeted alterations in the HIF-1α gene. Mutants demonstrated decreased survival and reproductive output and down-regulated genes for the HIF-1α-mediated pathway in low-oxygen conditions. These findings suggest that the HIF-1α pathway is a critical component of resistance to hypoxia in D. magna. This study provides novel insights into the molecular basis of hypoxia tolerance of HIF-1α in D. magna and expands our understanding of how aquatic organisms can adapt to or be challenged by changing oxygen levels in the face of global environmental changes.
Collapse
Affiliation(s)
- Yoseop Lee
- Department of Biological Sciences, College of Science, Sungkyunkwan University, Suwon 16419, South Korea
| | - Eunjin Byeon
- Department of Biological Sciences, College of Science, Sungkyunkwan University, Suwon 16419, South Korea
| | - Jin-Sol Lee
- Department of Biological Sciences, College of Science, Sungkyunkwan University, Suwon 16419, South Korea
| | - Piotr Maszczyk
- Department of Hydrobiology, Faculty of Biology, University of Warsaw, Warsaw, Poland
| | - Hyung Sik Kim
- School of Pharmacy, Sungkyunkwan University, Suwon 16419, South Korea
| | - Alaa El-Din H Sayed
- Department of Zoology, Faculty of Science, Assiut University, Assiut 71516, Egypt
| | - Zhou Yang
- Jiangsu Key Laboratory for Biodiversity and Biotechnology, School of Biological Sciences, Nanjing Normal University, Nanjing 210023, China
| | - Jae-Seong Lee
- Department of Biological Sciences, College of Science, Sungkyunkwan University, Suwon 16419, South Korea.
| | - Duck-Hyun Kim
- Department of Biological Sciences, College of Science, Sungkyunkwan University, Suwon 16419, South Korea.
| |
Collapse
|
4
|
Menegakis A, Vennin C, Ient J, Groot AJ, Krenning L, Klompmaker R, Friskes A, Ilic M, Yaromina A, Harkes R, van den Broek B, Jakob Sonke J, De Jong M, Piepers J, van Rheenen J, Vooijs MA, Medema RH. A novel lineage-tracing tool reveals that hypoxic tumor cells drive tumor relapse after radiotherapy. Radiother Oncol 2024:110592. [PMID: 39427933 DOI: 10.1016/j.radonc.2024.110592] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2024] [Revised: 09/19/2024] [Accepted: 09/24/2024] [Indexed: 10/22/2024]
Abstract
PURPOSE Tumor hypoxia imposes a main obstacle to the efficacy of anti-cancer therapy. Understanding the cellular dynamics of individual hypoxic cells before, during and post-treatment has been hampered by the technical inability to identify and trace these cells over time. METHODS AND MATERIALS Here, we present a novel lineage-tracing reporter for hypoxic cells based on the conditional expression of a HIF1a-CreERT2-UnaG biosensor that can visualize hypoxic cells in a time-dependent manner and trace the fate of hypoxic cells over time. We combine this system with multiphoton microscopy, flow cytometry, and immunofluorescence to characterize the role of hypoxic cells in tumor relapse after irradiation in H1299 tumor spheroids and in vivo xenografts. RESULTS We validate the reporter in monolayer cultures and we show that tagged cells colocalize in spheroids and human tumor xenografts with the hypoxic marker pimonidazole. We found that irradiation of H1299-HIFcreUnaG spheroids leads to preferential outgrowth of cells from the hypoxic core. Similarly, in xenografts tumors, although initially UnaG-positive-cells coincide with pimonidazole-positive tumor areas and they are merely quiescent, upon Irradiation UnaG-positive cells enrich in regrowing tumors and are mainly proliferative. CONCLUSIONS Collectively, our data provide clear evidence that the hypoxic cells drive tumor relapse after irradiation.
Collapse
Affiliation(s)
- Apostolos Menegakis
- Oncode Institute, Division of Cell Biology, The Netherlands Cancer Institute, Plesmanlaan 121, 1066 CX, Amsterdam, the Netherland; Bioimaging Facility, The Netherlands Cancer Institute, Plesmanlaan 121, 1066 CX, Amsterdam, the Netherlands.
| | - Claire Vennin
- Division of Molecular Pathology, The Netherlands Cancer Institute, Plesmanlaan 121, 1066 CX, Amsterdam, the Netherlands
| | - Jonathan Ient
- Department of Radiation Oncology (Maastro), GROW School for Oncology and Reproduction, Maastricht University Medical Centre, 6200 MD Maastricht, the Netherlands
| | - Arjan J Groot
- Department of Radiation Oncology (Maastro), GROW School for Oncology and Reproduction, Maastricht University Medical Centre, 6200 MD Maastricht, the Netherlands
| | - Lenno Krenning
- Oncode Institute, Division of Cell Biology, The Netherlands Cancer Institute, Plesmanlaan 121, 1066 CX, Amsterdam, the Netherland
| | - Rob Klompmaker
- Oncode Institute, Division of Cell Biology, The Netherlands Cancer Institute, Plesmanlaan 121, 1066 CX, Amsterdam, the Netherland
| | - Anoek Friskes
- Oncode Institute, Division of Cell Biology, The Netherlands Cancer Institute, Plesmanlaan 121, 1066 CX, Amsterdam, the Netherland
| | - Mila Ilic
- Oncode Institute, Division of Cell Biology, The Netherlands Cancer Institute, Plesmanlaan 121, 1066 CX, Amsterdam, the Netherland
| | - Ala Yaromina
- The M-Lab, Department of Precision Medicine, GROW - School for Oncology and Reproduction, Maastricht University, Maastricht, the Netherlands
| | - Rolf Harkes
- Bioimaging Facility, The Netherlands Cancer Institute, Plesmanlaan 121, 1066 CX, Amsterdam, the Netherlands
| | - Bram van den Broek
- Bioimaging Facility, The Netherlands Cancer Institute, Plesmanlaan 121, 1066 CX, Amsterdam, the Netherlands
| | - Jan Jakob Sonke
- Department of Radiation Oncology, The Netherlands Cancer Institute, Plesmanlaan 121, 1066 CX, Amsterdam, the Netherlands
| | - Monique De Jong
- Department of Radiation Oncology, The Netherlands Cancer Institute, Plesmanlaan 121, 1066 CX, Amsterdam, the Netherlands
| | - Jolanda Piepers
- Department of Radiation Oncology (Maastro), GROW School for Oncology and Reproduction, Maastricht University Medical Centre, 6200 MD Maastricht, the Netherlands
| | - Jacco van Rheenen
- Division of Molecular Pathology, The Netherlands Cancer Institute, Plesmanlaan 121, 1066 CX, Amsterdam, the Netherlands
| | - Marc A Vooijs
- Department of Radiation Oncology (Maastro), GROW School for Oncology and Reproduction, Maastricht University Medical Centre, 6200 MD Maastricht, the Netherlands.
| | - René H Medema
- Oncode Institute, Division of Cell Biology, The Netherlands Cancer Institute, Plesmanlaan 121, 1066 CX, Amsterdam, the Netherland; Princess Maxima Center for Pediatric Oncology, Utrecht, the Netherlands(2).
| |
Collapse
|
5
|
Chatzitheodoridou D, Bureik D, Padovani F, Nadimpalli KV, Schmoller KM. Decoupled transcript and protein concentrations ensure histone homeostasis in different nutrients. EMBO J 2024:10.1038/s44318-024-00227-w. [PMID: 39271795 DOI: 10.1038/s44318-024-00227-w] [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: 02/07/2024] [Revised: 08/05/2024] [Accepted: 08/27/2024] [Indexed: 09/15/2024] Open
Abstract
To maintain protein homeostasis in changing nutrient environments, cells must precisely control the amount of their proteins, despite the accompanying changes in cell growth and biosynthetic capacity. As nutrients are major regulators of cell cycle length and progression, a particular challenge arises for the nutrient-dependent regulation of 'cell cycle genes', which are periodically expressed during the cell cycle. One important example are histones, which are needed at a constant histone-to-DNA stoichiometry. Here we show that budding yeast achieves histone homeostasis in different nutrients through a decoupling of transcript and protein abundance. We find that cells downregulate histone transcripts in poor nutrients to avoid toxic histone overexpression, but produce constant amounts of histone proteins through nutrient-specific regulation of translation efficiency. Our findings suggest that this allows cells to balance the need for rapid histone production under fast growth conditions with the tight regulation required to avoid toxic overexpression in poor nutrients.
Collapse
Affiliation(s)
- Dimitra Chatzitheodoridou
- Institute of Functional Epigenetics, Molecular Targets and Therapeutics Center, Helmholtz Zentrum München, 85764, Neuherberg, Germany
| | - Daniela Bureik
- Institute of Functional Epigenetics, Molecular Targets and Therapeutics Center, Helmholtz Zentrum München, 85764, Neuherberg, Germany
| | - Francesco Padovani
- Institute of Functional Epigenetics, Molecular Targets and Therapeutics Center, Helmholtz Zentrum München, 85764, Neuherberg, Germany
| | - Kalyan V Nadimpalli
- Institute of Functional Epigenetics, Molecular Targets and Therapeutics Center, Helmholtz Zentrum München, 85764, Neuherberg, Germany
| | - Kurt M Schmoller
- Institute of Functional Epigenetics, Molecular Targets and Therapeutics Center, Helmholtz Zentrum München, 85764, Neuherberg, Germany.
| |
Collapse
|
6
|
Ghaffari-Bohlouli P, Jafari H, Okoro OV, Alimoradi H, Nie L, Jiang G, Kakkar A, Shavandi A. Gas Therapy: Generating, Delivery, and Biomedical Applications. SMALL METHODS 2024; 8:e2301349. [PMID: 38193272 DOI: 10.1002/smtd.202301349] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/05/2023] [Revised: 12/11/2023] [Indexed: 01/10/2024]
Abstract
Oxygen (O2), nitric oxide (NO), carbon monoxide (CO), hydrogen sulfide (H2S), and hydrogen (H2) with direct effects, and carbon dioxide (CO2) with complementary effects on the condition of various diseases are known as therapeutic gases. The targeted delivery and in situ generation of these therapeutic gases with controllable release at the site of disease has attracted attention to avoid the risk of gas poisoning and improve their performance in treating various diseases such as cancer therapy, cardiovascular therapy, bone tissue engineering, and wound healing. Stimuli-responsive gas-generating sources and delivery systems based on biomaterials that enable on-demand and controllable release are promising approaches for precise gas therapy. This work highlights current advances in the design and development of new approaches and systems to generate and deliver therapeutic gases at the site of disease with on-demand release behavior. The performance of the delivered gases in various biomedical applications is then discussed.
Collapse
Affiliation(s)
- Pejman Ghaffari-Bohlouli
- 3BIO-BioMatter, École polytechnique de Bruxelles, Université Libre de Bruxelles (ULB), Avenue F.D. Roosevelt, 50-CP 165/61, Brussels, 1050, Belgium
- Department of Chemistry, McGill University, 801 Sherbrooke Street West, Montréal, Québec, H3A 0B8, Canada
| | - Hafez Jafari
- 3BIO-BioMatter, École polytechnique de Bruxelles, Université Libre de Bruxelles (ULB), Avenue F.D. Roosevelt, 50-CP 165/61, Brussels, 1050, Belgium
| | - Oseweuba Valentine Okoro
- 3BIO-BioMatter, École polytechnique de Bruxelles, Université Libre de Bruxelles (ULB), Avenue F.D. Roosevelt, 50-CP 165/61, Brussels, 1050, Belgium
| | - Houman Alimoradi
- 3BIO-BioMatter, École polytechnique de Bruxelles, Université Libre de Bruxelles (ULB), Avenue F.D. Roosevelt, 50-CP 165/61, Brussels, 1050, Belgium
| | - Lei Nie
- 3BIO-BioMatter, École polytechnique de Bruxelles, Université Libre de Bruxelles (ULB), Avenue F.D. Roosevelt, 50-CP 165/61, Brussels, 1050, Belgium
- College of Life Sciences, Xinyang Normal University, Xinyang, 464000, China
| | - Guohua Jiang
- School of Materials Science and Engineering, Zhejiang Sci-Tech University, Hangzhou, 310018, China
| | - Ashok Kakkar
- Department of Chemistry, McGill University, 801 Sherbrooke Street West, Montréal, Québec, H3A 0B8, Canada
| | - Amin Shavandi
- 3BIO-BioMatter, École polytechnique de Bruxelles, Université Libre de Bruxelles (ULB), Avenue F.D. Roosevelt, 50-CP 165/61, Brussels, 1050, Belgium
| |
Collapse
|
7
|
Meng B, Zhao N, Mlcochova P, Ferreira IATM, Ortmann BM, Davis T, Wit N, Rehwinkel J, Cook S, Maxwell PH, Nathan JA, Gupta RK. Hypoxia drives HIF2-dependent reversible macrophage cell cycle entry. Cell Rep 2024; 43:114471. [PMID: 38996069 DOI: 10.1016/j.celrep.2024.114471] [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: 03/17/2024] [Revised: 05/22/2024] [Accepted: 06/24/2024] [Indexed: 07/14/2024] Open
Abstract
Low-oxygen conditions (hypoxia) have been associated primarily with cell-cycle arrest in dividing cells. Macrophages are typically quiescent in G0 but can proliferate in response to tissue signals. Here we show that hypoxia (1% oxygen tension) results in reversible entry into the cell cycle in macrophages. Cell cycle progression is largely limited to G0-G1/S phase transition with little progression to G2/M. This cell cycle transitioning is triggered by an HIF2α-directed transcriptional program. The response is accompanied by increased expression of cell-cycle-associated proteins, including CDK1, which is known to phosphorylate SAMHD1 at T592 and thereby regulate antiviral activity. Prolyl hydroxylase (PHD) inhibitors are able to recapitulate HIF2α-dependent cell cycle entry in macrophages. Finally, tumor-associated macrophages (TAMs) in lung cancers exhibit transcriptomic profiles representing responses to low oxygen and cell cycle progression at the single-cell level. These findings have implications for inflammation and tumor progression/metastasis where low-oxygen environments are common.
Collapse
Affiliation(s)
- Bo Meng
- Cambridge Institute of Therapeutic Immunology and Infectious Disease (CITIID), Cambridge, UK; Department of Medicine, University of Cambridge, Cambridge, UK.
| | - Na Zhao
- University of Oxford, Oxford, UK
| | - Petra Mlcochova
- Cambridge Institute of Therapeutic Immunology and Infectious Disease (CITIID), Cambridge, UK; Department of Medicine, University of Cambridge, Cambridge, UK
| | - Isabella A T M Ferreira
- Cambridge Institute of Therapeutic Immunology and Infectious Disease (CITIID), Cambridge, UK; Department of Medicine, University of Cambridge, Cambridge, UK
| | - Brian M Ortmann
- Cambridge Institute of Therapeutic Immunology and Infectious Disease (CITIID), Cambridge, UK; Department of Medicine, University of Cambridge, Cambridge, UK
| | | | - Niek Wit
- Cambridge Institute of Therapeutic Immunology and Infectious Disease (CITIID), Cambridge, UK; Department of Medicine, University of Cambridge, Cambridge, UK
| | | | | | | | - James A Nathan
- Cambridge Institute of Therapeutic Immunology and Infectious Disease (CITIID), Cambridge, UK; Department of Medicine, University of Cambridge, Cambridge, UK
| | - Ravindra K Gupta
- Cambridge Institute of Therapeutic Immunology and Infectious Disease (CITIID), Cambridge, UK; Department of Medicine, University of Cambridge, Cambridge, UK; Africa Health Research Institute, Durban, KwaZulu Natal, South Africa.
| |
Collapse
|
8
|
Laird M, Ku JC, Raiten J, Sriram S, Moore M, Li Y. Mitochondrial metabolism regulation and epigenetics in hypoxia. Front Physiol 2024; 15:1393232. [PMID: 38915781 PMCID: PMC11194441 DOI: 10.3389/fphys.2024.1393232] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2024] [Accepted: 05/13/2024] [Indexed: 06/26/2024] Open
Abstract
The complex and dynamic interaction between cellular energy control and gene expression modulation is shown by the intersection between mitochondrial metabolism and epigenetics in hypoxic environments. Poor oxygen delivery to tissues, or hypoxia, is a basic physiological stressor that sets off a series of reactions in cells to adapt and endure oxygen-starved environments. Often called the "powerhouse of the cell," mitochondria are essential to cellular metabolism, especially regarding producing energy through oxidative phosphorylation. The cellular response to hypoxia entails a change in mitochondrial metabolism to improve survival, including epigenetic modifications that control gene expression without altering the underlying genome. By altering the expression of genes involved in angiogenesis, cell survival, and metabolism, these epigenetic modifications help cells adapt to hypoxia. The sophisticated interplay between mitochondrial metabolism and epigenetics in hypoxia is highlighted by several important points, which have been summarized in the current article. Deciphering the relationship between mitochondrial metabolism and epigenetics during hypoxia is essential to understanding the molecular processes that regulate cellular adaptation to reduced oxygen concentrations.
Collapse
Affiliation(s)
- Madison Laird
- Western Michigan University Homer Stryker School of Medicine, Kalamazoo, MI, United States
| | - Jennifer C. Ku
- Western Michigan University Homer Stryker School of Medicine, Kalamazoo, MI, United States
| | - Jacob Raiten
- Western Michigan University Homer Stryker School of Medicine, Kalamazoo, MI, United States
| | - Sashwat Sriram
- Western Michigan University Homer Stryker School of Medicine, Kalamazoo, MI, United States
| | - Megan Moore
- Western Michigan University Homer Stryker School of Medicine, Kalamazoo, MI, United States
| | - Yong Li
- Department of Orthopaedic Surgery, Biomedical Engineering, Western Michigan University Homer Stryker School of Medicine, Kalamazoo, MI, United States
| |
Collapse
|
9
|
Tanaka A, Ogawa M, Zhou Y, Namba K, Hendrickson RC, Miele MM, Li Z, Klimstra DS, Buckley PG, Gulcher J, Wang JY, Roehrl MHA. Proteogenomic characterization of primary colorectal cancer and metastatic progression identifies proteome-based subtypes and signatures. Cell Rep 2024; 43:113810. [PMID: 38377004 DOI: 10.1016/j.celrep.2024.113810] [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: 11/20/2022] [Revised: 10/26/2023] [Accepted: 02/01/2024] [Indexed: 02/22/2024] Open
Abstract
Metastatic progression of colorectal adenocarcinoma (CRC) remains poorly understood and poses significant challenges for treatment. To overcome these challenges, we performed multiomics analyses of primary CRC and liver metastases. Genomic alterations, such as structural variants or copy number alterations, were enriched in oncogenes and tumor suppressor genes and increased in metastases. Unsupervised mass spectrometry-based proteomics of 135 primary and 123 metastatic CRCs uncovered distinct proteomic subtypes, three each for primary and metastatic CRCs, respectively. Integrated analyses revealed that hypoxia, stemness, and immune signatures characterize these 6 subtypes. Hypoxic CRC harbors high epithelial-to-mesenchymal transition features and metabolic adaptation. CRC with a stemness signature shows high oncogenic pathway activation and alternative telomere lengthening (ALT) phenotype, especially in metastatic lesions. Tumor microenvironment analysis shows immune evasion via modulation of major histocompatibility complex (MHC) class I/II and antigen processing pathways. This study characterizes both primary and metastatic CRCs and provides a large proteogenomics dataset of metastatic progression.
Collapse
Affiliation(s)
- Atsushi Tanaka
- Department of Pathology, Beth Israel Deaconess Medical Center, Boston, MA, USA; Harvard Medical School, Boston, MA, USA; Department of Pathology and Laboratory Medicine, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Makiko Ogawa
- Department of Pathology, Beth Israel Deaconess Medical Center, Boston, MA, USA; Harvard Medical School, Boston, MA, USA; Department of Pathology and Laboratory Medicine, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Yihua Zhou
- Department of Pathology and Laboratory Medicine, Memorial Sloan Kettering Cancer Center, New York, NY, USA; ICU Department, Second Affiliated Hospital of Nanchang University, Nanchang, Jiangxi, China
| | - Kei Namba
- Department of Pathology and Laboratory Medicine, Memorial Sloan Kettering Cancer Center, New York, NY, USA; Department of General Thoracic Surgery and Breast and Endocrinological Surgery, Okayama University Graduate School of Medicine, Dentistry, and Pharmaceutical Sciences, Okayama, Japan
| | - Ronald C Hendrickson
- Sloan Kettering Institute, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Matthew M Miele
- Sloan Kettering Institute, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Zhuoning Li
- Sloan Kettering Institute, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - David S Klimstra
- Department of Pathology and Laboratory Medicine, Memorial Sloan Kettering Cancer Center, New York, NY, USA; Paige.AI, New York, NY, USA
| | | | | | | | - Michael H A Roehrl
- Department of Pathology, Beth Israel Deaconess Medical Center, Boston, MA, USA; Harvard Medical School, Boston, MA, USA; Department of Pathology and Laboratory Medicine, Memorial Sloan Kettering Cancer Center, New York, NY, USA.
| |
Collapse
|
10
|
Arseni C, Samiotaki M, Panayotou G, Simos G, Mylonis I. Combinatorial regulation by ERK1/2 and CK1δ protein kinases leads to HIF-1α association with microtubules and facilitates its symmetrical distribution during mitosis. Cell Mol Life Sci 2024; 81:72. [PMID: 38300329 PMCID: PMC10834586 DOI: 10.1007/s00018-024-05120-7] [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: 10/30/2023] [Revised: 12/13/2023] [Accepted: 01/07/2024] [Indexed: 02/02/2024]
Abstract
Hypoxia-inducible factor-1 (HIF-1) is the key transcriptional mediator of the cellular response to hypoxia and is also involved in cancer progression. Regulation of its oxygen-sensitive HIF-1α subunit involves post-translational modifications that control its stability, subcellular localization, and activity. We have previously reported that phosphorylation of the HIF-1α C-terminal domain by ERK1/2 promotes HIF-1α nuclear accumulation and stimulates HIF-1 activity while lack of this modification triggers HIF-1α nuclear export and its association with mitochondria. On the other hand, modification of the N-terminal domain of HIF-1α by CK1δ impairs HIF-1 activity by obstructing the formation of a HIF-1α/ARNT heterodimer. Investigation of these two antagonistic events by expressing double phospho-site mutants in HIF1A-/- cells under hypoxia revealed independent and additive phosphorylation effects that can create a gradient of HIF-1α subcellular localization and transcriptional activity. Furthermore, modification by CK1δ caused mitochondrial release of the non-nuclear HIF-1α form and binding to microtubules via its N-terminal domain. In agreement, endogenous HIF-1α could be shown to co-localize with mitotic spindle microtubules and interact with tubulin, both of which were inhibited by CK1δ silencing or inhibition. Moreover, CK1δ expression was necessary for equal partitioning of mother cell-produced HIF-1α to the daughter cell nuclei at the end of mitosis. Overall, our results suggest that phosphorylation by CK1δ stimulates the association of non-nuclear HIF-1α with microtubules, which may serve as a means to establish a symmetric distribution of HIF-1α during cell division under low oxygen conditions.
Collapse
Affiliation(s)
- Christina Arseni
- Laboratory of Biochemistry, Faculty of Medicine, University of Thessaly, 41500, Larissa, Greece
| | - Martina Samiotaki
- Institute for Bio-Innovation, BSRC "Alexander Fleming", 16672, Vari, Greece
| | - George Panayotou
- Institute for Bio-Innovation, BSRC "Alexander Fleming", 16672, Vari, Greece
| | - George Simos
- Laboratory of Biochemistry, Faculty of Medicine, University of Thessaly, 41500, Larissa, Greece.
- Gerald Bronfman Department of Oncology, Faculty of Medicine, McGill University, Montreal, Canada.
| | - Ilias Mylonis
- Laboratory of Biochemistry, Faculty of Medicine, University of Thessaly, 41500, Larissa, Greece.
| |
Collapse
|
11
|
Louise Kelly C, Wydrzynska M, Phelan MM, Osharovich S, Delikatny EJ, Sée V, Poptani H. Inhibition of glioblastoma cell proliferation and invasion by the choline-kinase inhibitor JAS239 varies with cell type and hypoxia. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.01.17.576078. [PMID: 38293093 PMCID: PMC10827177 DOI: 10.1101/2024.01.17.576078] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/01/2024]
Abstract
Background Elevated choline kinase alpha (ChoK) is observed in most solid tumours including glioblastomas (GBM), yet until recently, inhibitors of ChoK have demonstrated limited efficacy in GBM models. Given that hypoxia is associated with GBM therapy resistance, we hypothesised that tumour hypoxia could be responsible for such limitations. We therefore evaluated in GBM cells, the effect of hypoxia on the function of JAS239, a potent ChoK inhibitor. Methods Rodent (F98 and 9L) and human (U-87 MG and U-251 MG) GBM cell lines were subjected to 72 hours of hypoxia conditioning and treated with JAS239 for 24 hours. NMR metabolomic measurements and analyses were performed to evaluate the signalling pathways involved. In addition, cell proliferation, cell cycle progression and cell invasion were measured in cell monolayers and 3D spheroids, with or without JAS239 treatment in normoxic or hypoxic cells to assess how hypoxia affects JAS239 function. Results Hypoxia and JAS239 treatment led to significant changes in the cellular metabolic pathways, specifically the phospholipid and glycolytic pathways associated with a reduction in cell proliferation via induced cell cycle arrest. Interestingly, JAS239 also impaired GBM invasion. However, JAS239 effects were variable depending on the cell line, reflecting the inherent heterogeneity observed in GBMs. Conclusion Our findings indicate that JAS239 and hypoxia can deregulate cellular metabolism, inhibit proliferation and alter cell invasion. These results may be useful for the design of new therapeutic strategies based on ChoK inhibition that can act on multiple pro-tumorigenic features.
Collapse
Affiliation(s)
- Claire Louise Kelly
- Centre for Preclinical Imaging, Department of Molecular and Clinical Cancer Medicine, University of Liverpool, Liverpool, UK
- Centre for Cell Imaging, Department of Biochemistry & Systems Biology, University of Liverpool, Liverpool, UK
| | - Martyna Wydrzynska
- Centre for Cell Imaging, Department of Biochemistry & Systems Biology, University of Liverpool, Liverpool, UK
| | - Marie M Phelan
- High field NMR facility, Department of Biochemistry & Systems Biology, University of Liverpool, UK
| | - Sofya Osharovich
- Department of Radiology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, United States of America
| | - Edward J. Delikatny
- Department of Radiology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, United States of America
| | - Violaine Sée
- Centre for Cell Imaging, Department of Biochemistry & Systems Biology, University of Liverpool, Liverpool, UK
| | - Harish Poptani
- Centre for Preclinical Imaging, Department of Molecular and Clinical Cancer Medicine, University of Liverpool, Liverpool, UK
| |
Collapse
|
12
|
Nisar H, Labonté FM, Roggan MD, Schmitz C, Chevalier F, Konda B, Diegeler S, Baumstark-Khan C, Hellweg CE. Hypoxia Modulates Radiosensitivity and Response to Different Radiation Qualities in A549 Non-Small Cell Lung Cancer (NSCLC) Cells. Int J Mol Sci 2024; 25:1010. [PMID: 38256084 PMCID: PMC10816011 DOI: 10.3390/ijms25021010] [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: 11/23/2023] [Revised: 12/28/2023] [Accepted: 01/09/2024] [Indexed: 01/24/2024] Open
Abstract
Hypoxia-induced radioresistance reduces the efficacy of radiotherapy for solid malignancies, including non-small cell lung cancer (NSCLC). Cellular hypoxia can confer radioresistance through cellular and tumor micro-environment adaptations. Until recently, studies evaluating radioresistance secondary to hypoxia were designed to maintain cellular hypoxia only before and during irradiation, while any handling of post-irradiated cells was carried out in standard oxic conditions due to the unavailability of hypoxia workstations. This limited the possibility of simulating in vivo or clinical conditions in vitro. The presence of molecular oxygen is more important for the radiotoxicity of low-linear energy transfer (LET) radiation (e.g., X-rays) than that of high-LET carbon (12C) ions. The mechanisms responsible for 12C ions' potential to overcome hypoxia-induced radioresistance are currently not fully understood. Therefore, the radioresistance of hypoxic A549 NSCLC cells following exposure to X-rays or 12C ions was investigated along with cell cycle progression and gene expression by maintaining hypoxia before, during and after irradiation. A549 cells were incubated under normoxia (20% O2) or hypoxia (1% O2) for 48 h and then irradiated with X-rays (200 kV) or 12C ions (35 MeV/n, LET ~75 keV/µm). Cell survival was evaluated using colony-forming ability (CFA) assays immediately or 24 h after irradiation (late plating). DNA double-strand breaks (DSBs) were analyzed using γH2AX immunofluorescence microscopy. Cell cycle progression was determined by flow cytometry of 4',6-diamidino-2-phenylindole-stained cells. The global transcription profile post-irradiation was evaluated by RNA sequencing. When hypoxia was maintained before, during and after irradiation, hypoxia-induced radioresistance was observed only in late plating CFA experiments. The killing efficiency of 12C ions was much higher than that of X-rays. Cell survival under hypoxia was affected more strongly by the timepoint of plating in the case of X-rays compared to 12C ions. Cell cycle arrest following irradiation under hypoxia was less pronounced but more prolonged. DSB induction and resolution following irradiation were not significantly different under normoxia and hypoxia. Gene expression response to irradiation primarily comprised cell cycle regulation for both radiation qualities and oxygen conditions. Several PI3K target genes involved in cell migration and cell motility were differentially upregulated in hypoxic cells. Hypoxia-induced radioresistance may be linked to altered cell cycle response to irradiation and PI3K-mediated changes in cell motility and migration in A549 cells rather than less DNA damage or faster repair.
Collapse
Affiliation(s)
- Hasan Nisar
- Department of Radiation Biology, Institute of Aerospace Medicine, German Aerospace Center (DLR), 51147 Cologne, Germany; (H.N.); (F.M.L.); (M.D.R.); (C.S.); (B.K.); (S.D.); (C.B.-K.)
- Department of Medical Sciences, Pakistan Institute of Engineering and Applied Sciences (PIEAS), Islamabad 44000, Pakistan
| | - Frederik M. Labonté
- Department of Radiation Biology, Institute of Aerospace Medicine, German Aerospace Center (DLR), 51147 Cologne, Germany; (H.N.); (F.M.L.); (M.D.R.); (C.S.); (B.K.); (S.D.); (C.B.-K.)
- Center for Molecular Medicine Cologne (CMMC), University of Cologne, 50931 Cologne, Germany
| | - Marie Denise Roggan
- Department of Radiation Biology, Institute of Aerospace Medicine, German Aerospace Center (DLR), 51147 Cologne, Germany; (H.N.); (F.M.L.); (M.D.R.); (C.S.); (B.K.); (S.D.); (C.B.-K.)
- German Center for Neurodegenerative Diseases (DZNE), 53127 Bonn, Germany
| | - Claudia Schmitz
- Department of Radiation Biology, Institute of Aerospace Medicine, German Aerospace Center (DLR), 51147 Cologne, Germany; (H.N.); (F.M.L.); (M.D.R.); (C.S.); (B.K.); (S.D.); (C.B.-K.)
| | - François Chevalier
- UMR6252 CIMAP, CEA-CNRS-ENSICAEN-University of Caen Normandy, 14000 Caen, France;
| | - Bikash Konda
- Department of Radiation Biology, Institute of Aerospace Medicine, German Aerospace Center (DLR), 51147 Cologne, Germany; (H.N.); (F.M.L.); (M.D.R.); (C.S.); (B.K.); (S.D.); (C.B.-K.)
| | - Sebastian Diegeler
- Department of Radiation Biology, Institute of Aerospace Medicine, German Aerospace Center (DLR), 51147 Cologne, Germany; (H.N.); (F.M.L.); (M.D.R.); (C.S.); (B.K.); (S.D.); (C.B.-K.)
- Department of Radiation Oncology, UT Southwestern Medical Center, Dallas, TX 75390, USA
| | - Christa Baumstark-Khan
- Department of Radiation Biology, Institute of Aerospace Medicine, German Aerospace Center (DLR), 51147 Cologne, Germany; (H.N.); (F.M.L.); (M.D.R.); (C.S.); (B.K.); (S.D.); (C.B.-K.)
| | - Christine E. Hellweg
- Department of Radiation Biology, Institute of Aerospace Medicine, German Aerospace Center (DLR), 51147 Cologne, Germany; (H.N.); (F.M.L.); (M.D.R.); (C.S.); (B.K.); (S.D.); (C.B.-K.)
| |
Collapse
|
13
|
Zhu M, Hu W, Lin L, Yang Q, Zhang L, Xu J, Xu Y, Liu J, Zhang M, Tong X, Zhu K, Feng K, Feng Y, Su J, Huang X, Li J. Single-cell RNA sequencing reveals new subtypes of lens superficial tissue in humans. Cell Prolif 2023; 56:e13477. [PMID: 37057399 PMCID: PMC10623935 DOI: 10.1111/cpr.13477] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2023] [Revised: 03/27/2023] [Accepted: 04/01/2023] [Indexed: 04/15/2023] Open
Abstract
Although the cell atlas of the human ocular anterior segment of the human eye was revealed by single-nucleus RNA sequencing, whether subtypes of lens stem/progenitor cells exist among epithelial cells and the molecular characteristics of cell differentiation of the human lens remain unclear. Single-cell RNA sequencing is a powerful tool to analyse the heterogeneity of tissues at the single cell level, leading to a better understanding of the processes of cell differentiation. By profiling 18,596 cells in human lens superficial tissue through single-cell sequencing, we identified two subtypes of lens epithelial cells that specifically expressed C8orf4 and ADAMTSL4 with distinct spatial localization, a new type of fibre cells located directly adjacent to the epithelium, and a subpopulation of ADAMTSL4+ cells that might be lens epithelial stem/progenitor cells. We also found two trajectories of lens epithelial cell differentiation and changes of some important genes during differentiation.
Collapse
Affiliation(s)
- Meng‐Chao Zhu
- State Key Laboratory of Ophthalmology, Optometry and Visual Science, Eye HospitalWenzhou Medical UniversityWenzhouChina
- National Clinical Research Center for Ocular Diseases, Eye HospitalWenzhou Medical UniversityWenzhouChina
| | - Wei Hu
- Department of Integrative Medicine and Neurobiology, School of Basic Medical Sciences, Institutes of Brain Science, Brain Science Collaborative Innovation Center, State Key Laboratory of Medical Neurobiology, Institute of Acupuncture and Moxibustion, Fudan Institutes of Integrative MedicineFudan UniversityShanghaiChina
| | - Lei Lin
- State Key Laboratory of Ophthalmology, Optometry and Visual Science, Eye HospitalWenzhou Medical UniversityWenzhouChina
- National Clinical Research Center for Ocular Diseases, Eye HospitalWenzhou Medical UniversityWenzhouChina
| | - Qing‐Wen Yang
- State Key Laboratory of Ophthalmology, Optometry and Visual Science, Eye HospitalWenzhou Medical UniversityWenzhouChina
- National Clinical Research Center for Ocular Diseases, Eye HospitalWenzhou Medical UniversityWenzhouChina
| | - Lu Zhang
- State Key Laboratory of Ophthalmology, Optometry and Visual Science, Eye HospitalWenzhou Medical UniversityWenzhouChina
- National Clinical Research Center for Ocular Diseases, Eye HospitalWenzhou Medical UniversityWenzhouChina
| | - Jia‐Lin Xu
- State Key Laboratory of Ophthalmology, Optometry and Visual Science, Eye HospitalWenzhou Medical UniversityWenzhouChina
- National Clinical Research Center for Ocular Diseases, Eye HospitalWenzhou Medical UniversityWenzhouChina
| | - Yi‐Tong Xu
- State Key Laboratory of Ophthalmology, Optometry and Visual Science, Eye HospitalWenzhou Medical UniversityWenzhouChina
- National Clinical Research Center for Ocular Diseases, Eye HospitalWenzhou Medical UniversityWenzhouChina
| | - Jia‐Sheng Liu
- State Key Laboratory of Ophthalmology, Optometry and Visual Science, Eye HospitalWenzhou Medical UniversityWenzhouChina
- National Clinical Research Center for Ocular Diseases, Eye HospitalWenzhou Medical UniversityWenzhouChina
| | - Meng‐Di Zhang
- State Key Laboratory of Ophthalmology, Optometry and Visual Science, Eye HospitalWenzhou Medical UniversityWenzhouChina
- National Clinical Research Center for Ocular Diseases, Eye HospitalWenzhou Medical UniversityWenzhouChina
| | - Xiao‐Yu Tong
- Zhejiang Provincial Clinical Research Center for Pediatric DiseaseThe Second Affiliated Hospital of Wenzhou Medical UniversityWenzhouZhejiangChina
| | - Kai‐Yi Zhu
- State Key Laboratory of Ophthalmology, Optometry and Visual Science, Eye HospitalWenzhou Medical UniversityWenzhouChina
- National Clinical Research Center for Ocular Diseases, Eye HospitalWenzhou Medical UniversityWenzhouChina
| | - Ke Feng
- State Key Laboratory of Ophthalmology, Optometry and Visual Science, Eye HospitalWenzhou Medical UniversityWenzhouChina
- National Clinical Research Center for Ocular Diseases, Eye HospitalWenzhou Medical UniversityWenzhouChina
| | - Yi Feng
- Department of Integrative Medicine and Neurobiology, School of Basic Medical Sciences, Institutes of Brain Science, Brain Science Collaborative Innovation Center, State Key Laboratory of Medical Neurobiology, Institute of Acupuncture and Moxibustion, Fudan Institutes of Integrative MedicineFudan UniversityShanghaiChina
| | - Jian‐Zhong Su
- State Key Laboratory of Ophthalmology, Optometry and Visual Science, Eye HospitalWenzhou Medical UniversityWenzhouChina
- National Clinical Research Center for Ocular Diseases, Eye HospitalWenzhou Medical UniversityWenzhouChina
| | - Xiu‐Feng Huang
- Zhejiang Provincial Clinical Research Center for Pediatric DiseaseThe Second Affiliated Hospital of Wenzhou Medical UniversityWenzhouZhejiangChina
| | - Jin Li
- State Key Laboratory of Ophthalmology, Optometry and Visual Science, Eye HospitalWenzhou Medical UniversityWenzhouChina
- National Clinical Research Center for Ocular Diseases, Eye HospitalWenzhou Medical UniversityWenzhouChina
| |
Collapse
|
14
|
Nisar H, Sanchidrián González PM, Brauny M, Labonté FM, Schmitz C, Roggan MD, Konda B, Hellweg CE. Hypoxia Changes Energy Metabolism and Growth Rate in Non-Small Cell Lung Cancer Cells. Cancers (Basel) 2023; 15:cancers15092472. [PMID: 37173939 PMCID: PMC10177580 DOI: 10.3390/cancers15092472] [Citation(s) in RCA: 7] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2023] [Revised: 04/18/2023] [Accepted: 04/19/2023] [Indexed: 05/15/2023] Open
Abstract
Hypoxia occurs in 80% of non-small cell lung carcinoma (NSCLC) cases, leading to treatment resistance. Hypoxia's effects on NSCLC energetics are not well-characterized. We evaluated changes in glucose uptake and lactate production in two NSCLC cell lines under hypoxia in conjunction with growth rate and cell cycle phase distribution. The cell lines A549 (p53 wt) and H358 (p53 null) were incubated under hypoxia (0.1% and 1% O2) or normoxia (20% O2). Glucose and lactate concentrations in supernatants were measured using luminescence assays. Growth kinetics were followed over seven days. Cell nuclei were stained with DAPI and nuclear DNA content was determined by flow cytometry to determine cell cycle phase. Gene expression under hypoxia was determined by RNA sequencing. Glucose uptake and lactate production under hypoxia were greater than under normoxia. They were also significantly greater in A549 compared to H358 cells. Faster energy metabolism in A549 cells was associated with a higher growth rate in comparison to H358 cells under both normoxia and hypoxia. In both cell lines, hypoxia significantly slowed down the growth rate compared to proliferation under normoxic conditions. Hypoxia led to redistribution of cells in the different cycle phases: cells in G1 increased and the G2 population decreased. Glucose uptake and lactate production increase under hypoxia in NSCLC cells indicated greater shunting of glucose into glycolysis rather than into oxidative phosphorylation compared to normoxia, making adenosine triphosphate (ATP) production less efficient. This may explain the redistribution of hypoxic cells in the G1 cell cycle phase and the time increase for cell doubling. Energy metabolism changes were more prominent in faster-growing A549 cells compared to slower-growing H358 cells, indicating possible roles for the p53 status and inherent growth rate of different cancer cells. In both cell lines, genes associated with cell motility, locomotion and migration were upregulated under chronic hypoxia, indicating a strong stimulus to escape hypoxic conditions.
Collapse
Affiliation(s)
- Hasan Nisar
- Department of Radiation Biology, Institute of Aerospace Medicine, German Aerospace Center (DLR), 51147 Cologne, Germany
- Department of Medical Sciences, Pakistan Institute of Engineering and Applied Sciences (PIEAS), Nilore, Islamabad 44000, Pakistan
| | | | - Melanie Brauny
- Department of Radiation Biology, Institute of Aerospace Medicine, German Aerospace Center (DLR), 51147 Cologne, Germany
- Interfaculty Institute of Microbiology and Infection Medicine, Faculty of Science/Faculty of Medicine, University of Tübingen, 72074 Tübingen, Germany
| | - Frederik M Labonté
- Department of Radiation Biology, Institute of Aerospace Medicine, German Aerospace Center (DLR), 51147 Cologne, Germany
- Department of Biology, Faculty of Mathematics and Natural Sciences, University of Cologne, 50923 Cologne, Germany
| | - Claudia Schmitz
- Department of Radiation Biology, Institute of Aerospace Medicine, German Aerospace Center (DLR), 51147 Cologne, Germany
| | - Marie Denise Roggan
- Department of Radiation Biology, Institute of Aerospace Medicine, German Aerospace Center (DLR), 51147 Cologne, Germany
- Deutsches Zentrum für Neurodegenerative Erkrankungen (DZNE), 53127 Bonn, Germany
| | - Bikash Konda
- Department of Radiation Biology, Institute of Aerospace Medicine, German Aerospace Center (DLR), 51147 Cologne, Germany
| | - Christine E Hellweg
- Department of Radiation Biology, Institute of Aerospace Medicine, German Aerospace Center (DLR), 51147 Cologne, Germany
| |
Collapse
|
15
|
Li H, Chen X, Zuo Z, Wang J, Guo Y. Identification and Characterization of Peptides from Bovine Collagen Hydrolysates that Promote Myogenic Cell Proliferation. JOURNAL OF AGRICULTURAL AND FOOD CHEMISTRY 2023; 71:4876-4889. [PMID: 36917229 DOI: 10.1021/acs.jafc.2c08929] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/18/2023]
Abstract
In this study, bovine collagen hydrolysate was purified via a series of chromatograms, and the peptides with the highest activity for promoting myoblast proliferation were identified by LC-MS-MS. It was demonstrated that the peptide GDAGPPGPAGPAGPPGPIG (hydroxylation) could promote C2C12 proliferation (+18.5% ± 0.04, P < 0.05). The certain peptide was capable of regulating the myogenic cell cycle and inhibiting myogenic cell apoptosis. By combining molecular docking, quantitative real-time PCR, and metabonomics, we suggested that the peptide GDAGPPGPAGPAGPPGPIG (hydroxylation) might bind to FGFR1 and affect the expression of genes downstream of FGFR1 and influence protein synthesis to promote myoblast proliferation. The above results showed that the peptides isolated in this study have the potential to alleviate sarcopenia in the elderly.
Collapse
Affiliation(s)
- Hanfeng Li
- Key Laboratory of Photochemical Conversion and Optoelectronic Materials, Technical Institute of Physics and Chemistry, Chinese Academy of Sciences, Beijing 100190, China
- School of Chemical Sciences, University of Chinese Academy of Sciences, Yuquan Road 19A, Beijing 100049, China
| | - Xin Chen
- Key Laboratory of Photochemical Conversion and Optoelectronic Materials, Technical Institute of Physics and Chemistry, Chinese Academy of Sciences, Beijing 100190, China
- School of Chemical Sciences, University of Chinese Academy of Sciences, Yuquan Road 19A, Beijing 100049, China
| | - Zhijie Zuo
- Key Laboratory of Photochemical Conversion and Optoelectronic Materials, Technical Institute of Physics and Chemistry, Chinese Academy of Sciences, Beijing 100190, China
- School of Chemical Sciences, University of Chinese Academy of Sciences, Yuquan Road 19A, Beijing 100049, China
| | - Jianing Wang
- Key Laboratory of Photochemical Conversion and Optoelectronic Materials, Technical Institute of Physics and Chemistry, Chinese Academy of Sciences, Beijing 100190, China
| | - Yanchuan Guo
- Key Laboratory of Photochemical Conversion and Optoelectronic Materials, Technical Institute of Physics and Chemistry, Chinese Academy of Sciences, Beijing 100190, China
- School of Chemical Sciences, University of Chinese Academy of Sciences, Yuquan Road 19A, Beijing 100049, China
| |
Collapse
|
16
|
Lerma Clavero A, Boqvist PL, Ingelshed K, Bosdotter C, Sedimbi S, Jiang L, Wermeling F, Vojtesek B, Lane DP, Kannan P. MDM2 inhibitors, nutlin-3a and navtemadelin, retain efficacy in human and mouse cancer cells cultured in hypoxia. Sci Rep 2023; 13:4583. [PMID: 36941277 PMCID: PMC10027891 DOI: 10.1038/s41598-023-31484-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2022] [Accepted: 03/13/2023] [Indexed: 03/23/2023] Open
Abstract
Activation of p53 by small molecule MDM2 inhibitors can induce cell cycle arrest or death in p53 wildtype cancer cells. However, cancer cells exposed to hypoxia can develop resistance to other small molecules, such as chemotherapies, that activate p53. Here, we evaluated whether hypoxia could render cancer cells insensitive to two MDM2 inhibitors with different potencies, nutlin-3a and navtemadlin. Inhibitor efficacy and potency were evaluated under short-term hypoxic conditions in human and mouse cancer cells expressing different p53 genotypes (wild-type, mutant, or null). Treatment of wild-type p53 cancer cells with MDM2 inhibitors reduced cell growth by > 75% in hypoxia through activation of the p53-p21 signaling pathway; no inhibitor-induced growth reduction was observed in hypoxic mutant or null p53 cells except at very high concentrations. The concentration of inhibitors needed to induce the maximal p53 response was not significantly different in hypoxia compared to normoxia. However, inhibitor efficacy varied by species and by cell line, with stronger effects at lower concentrations observed in human cell lines than in mouse cell lines grown as 2D and 3D cultures. Together, these results indicate that MDM2 inhibitors retain efficacy in hypoxia, suggesting they could be useful for targeting acutely hypoxic cancer cells.
Collapse
Affiliation(s)
- Ada Lerma Clavero
- Department of Microbiology, Tumor and Cell Biology, Karolinska Institutet, 171 77, Stockholm, Sweden
- Department of Medical Cell Biology, Uppsala University, 751 23, Uppsala, Sweden
| | - Paula Lafqvist Boqvist
- Department of Microbiology, Tumor and Cell Biology, Karolinska Institutet, 171 77, Stockholm, Sweden
| | - Katrine Ingelshed
- Department of Microbiology, Tumor and Cell Biology, Karolinska Institutet, 171 77, Stockholm, Sweden
| | - Cecilia Bosdotter
- Department of Microbiology, Tumor and Cell Biology, Karolinska Institutet, 171 77, Stockholm, Sweden
| | - Saikiran Sedimbi
- Department of Microbiology, Tumor and Cell Biology, Karolinska Institutet, 171 77, Stockholm, Sweden
- Moderna Therapeutics, 200 Technology Square, Cambridge, MA, 02139, USA
| | - Long Jiang
- Department of Medicine Solna, Center for Molecular Medicine, Karolinska University Hospital and Karolinska Institutet, 171 77, Stockholm, Sweden
| | - Fredrik Wermeling
- Department of Medicine Solna, Center for Molecular Medicine, Karolinska University Hospital and Karolinska Institutet, 171 77, Stockholm, Sweden
| | - Borivoj Vojtesek
- RECAMO, Masaryk Memorial Cancer Institute, 656 53, Brno, Czech Republic
| | - David P Lane
- Department of Microbiology, Tumor and Cell Biology, Karolinska Institutet, 171 77, Stockholm, Sweden.
| | - Pavitra Kannan
- Department of Microbiology, Tumor and Cell Biology, Karolinska Institutet, 171 77, Stockholm, Sweden.
| |
Collapse
|
17
|
Yu H, He Y, Zhang J, Zhang Z, Zhang X. Hepatic transcriptome analysis reveals the metabolic strategies of largemouth bass (Micropterus salmoides) under different dissolved oxygen condition. COMPARATIVE BIOCHEMISTRY AND PHYSIOLOGY. PART D, GENOMICS & PROTEOMICS 2023; 45:101032. [PMID: 36371883 DOI: 10.1016/j.cbd.2022.101032] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/15/2022] [Revised: 10/27/2022] [Accepted: 10/31/2022] [Indexed: 11/06/2022]
Abstract
Dissolved oxygen (DO) affects aquatic animals at a fundamental level so that the differences in its metabolic strategies under prolonged hypoxic conditions need an urgent exploration. In this experiment, largemouth bass (Micropterus salmoides) were chronically exposed (6 weeks) to severe hypoxia (S-HYP, DO: 2.0 ± 0.4 mg/L) and mild hypoxia (M-HYP, DO: 5.1 ± 0.4 mg/L). Compared to the control group (CON, DO:8.4 ± 0.4 mg/L), 1196 and 232 differentially expressed genes (DEGs) were obtained in S-HYP and M-HPY groups via transcriptome analysis, respectively. In S-HYP, lipolysis was promoted while anabolism was blocked. Meanwhile, significantly less fat droplet area was observed in the liver histology of S-HYP. Additionally, the cell cycle also responded to hypoxia, being blocked in the G1 phase with the suspension of DNA replication process. In M-HYP, the processing of protein in the endoplasmic reticulum and the synthesis of various aminoacyl t-RNA were inhibited, and a novel balance of the urea cycle might be established in the biosynthesis of arginine. The key DEGs involved in the above metabolic pathways, such as atgl, cpt1, arg1, etc., were validated by Q-PCR yielding results consistent with transcriptome data. This study indicates that the largemouth bass is prone to increase the proportion of lipid as an energy supply to adapt to the reprogramming of energy metabolism, while reducing the rate of cell proliferation to adapt to chronic severe hypoxia. This is also an undescribed observation in fish liver metabolism that largemouth bass may transform the synthesis and processing strategies of protein when exposed to chronic mild hypoxia.
Collapse
Affiliation(s)
- Haodong Yu
- College of Fisheries, Huazhong Agricultural University; Engineering Research Center of Green Development for Conventional Aquatic Biological Industry in the Yangtze River Economic Belt, Ministry of Education; Hubei Provincial Engineering Laboratory for Pond Aquaculture, Wuhan 430070, China; Hubei Hongshan Laboratory, Wuhan 430070, China
| | - Ya He
- College of Fisheries, Huazhong Agricultural University; Engineering Research Center of Green Development for Conventional Aquatic Biological Industry in the Yangtze River Economic Belt, Ministry of Education; Hubei Provincial Engineering Laboratory for Pond Aquaculture, Wuhan 430070, China; Hubei Hongshan Laboratory, Wuhan 430070, China
| | - Jinying Zhang
- College of Fisheries, Huazhong Agricultural University; Engineering Research Center of Green Development for Conventional Aquatic Biological Industry in the Yangtze River Economic Belt, Ministry of Education; Hubei Provincial Engineering Laboratory for Pond Aquaculture, Wuhan 430070, China; Hubei Hongshan Laboratory, Wuhan 430070, China
| | - Ziyi Zhang
- College of Fisheries, Huazhong Agricultural University; Engineering Research Center of Green Development for Conventional Aquatic Biological Industry in the Yangtze River Economic Belt, Ministry of Education; Hubei Provincial Engineering Laboratory for Pond Aquaculture, Wuhan 430070, China; Hubei Hongshan Laboratory, Wuhan 430070, China
| | - Xuezhen Zhang
- College of Fisheries, Huazhong Agricultural University; Engineering Research Center of Green Development for Conventional Aquatic Biological Industry in the Yangtze River Economic Belt, Ministry of Education; Hubei Provincial Engineering Laboratory for Pond Aquaculture, Wuhan 430070, China; Hubei Hongshan Laboratory, Wuhan 430070, China.
| |
Collapse
|
18
|
Nor Nazli NA, Muthuraju S, Ahmad F, Mohamed Yusoff AA, Jaafar H, Shamsuddin S, Abdullah JM. Characterisation of Primary Human Hippocampal Astrocyte Cell Culture Following Exposure to Hypoxia. Malays J Med Sci 2023; 30:92-106. [PMID: 36875187 PMCID: PMC9984107 DOI: 10.21315/mjms2023.30.1.8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2022] [Accepted: 07/01/2022] [Indexed: 03/05/2023] Open
Abstract
Background The present study aimed to understand the characterisation of human hippocampal astrocyte following hypoxia exposure. Based on the preliminary screening, 15 min was chosen as the time point and the cells were exposed to different oxygen percentages. Methods The Trypan blue viability assay used to examine cell death. Immunofluorescence assay, glial fibrillary acidic protein (GFAP) was used to portray the morphology of astrocytes. The hypoxia-inducible factor 1 (HIF-1) staining was performed to confirm hypoxia induced cell death and there was a dramatic expression of HIF-1α displayed in exposed astrocyte cells compared to the control. In molecular level, genes were chosen, such as glyceraldehyde 3-phosphate dehydrogenase (GAPDH), GFAP, HIF-1α and B-cell lymphoma 2 (Bcl-2) and ran the reverse transcription-polymerase chain reaction (RT-PCR). Results Microscope revealed a filamentous and clear nucleus appearance in a control whereas the rupture nuclei with no rigid structure of the cell were found in the 3% oxygen. The control and hypoxia cells were also stained with the annexin V-fluorescein isothiocyanate (annexin V-FITC). Fluorescence microscope reveals astrocyte cells after hypoxia showed higher expression of nuclei but not in control. Merging PI and FITC showed the differences of nuclei expression between the control and hypoxia. In the molecular analysis, there were significant changes of GFAP, HIF-1α and Bcl-2 in hypoxia exposed cells when compared to the control group. Conclusion Cells that were exposed to hypoxia (3% oxygen for 15 min) clearly showed damage. General view of human hippocampal astrocyte genomic response to hypoxia was obtained.
Collapse
Affiliation(s)
- Nurul Atikah Nor Nazli
- Department of Neurosciences, School of Medical Sciences, Universiti Sains Malaysia, Kelantan, Malaysia.,Brain and Behaviour Cluster, School of Medical Sciences, Universiti Sains Malaysia, Kelantan, Malaysia
| | - Sangu Muthuraju
- Department of Neurosciences, School of Medical Sciences, Universiti Sains Malaysia, Kelantan, Malaysia.,Brain and Behaviour Cluster, School of Medical Sciences, Universiti Sains Malaysia, Kelantan, Malaysia
| | - Farizan Ahmad
- Department of Neurosciences, School of Medical Sciences, Universiti Sains Malaysia, Kelantan, Malaysia.,Brain and Behaviour Cluster, School of Medical Sciences, Universiti Sains Malaysia, Kelantan, Malaysia
| | - Abdul Aziz Mohamed Yusoff
- Department of Neurosciences, School of Medical Sciences, Universiti Sains Malaysia, Kelantan, Malaysia.,Brain and Behaviour Cluster, School of Medical Sciences, Universiti Sains Malaysia, Kelantan, Malaysia
| | - Hasnan Jaafar
- Department of Pathology, School of Medical Sciences, Universiti Sains Malaysia, Kelantan, Malaysia
| | - Shaharum Shamsuddin
- Department of Biomedicine, School of Health Sciences, Universiti Sains Malaysia, Kelantan, Malaysia
| | - Jafri Malin Abdullah
- Department of Neurosciences, School of Medical Sciences, Universiti Sains Malaysia, Kelantan, Malaysia.,Brain and Behaviour Cluster, School of Medical Sciences, Universiti Sains Malaysia, Kelantan, Malaysia
| |
Collapse
|
19
|
Sasabe Y, Niitani M, Teramoto C, Yamaga S, Shime N, Tanabe K, Kataoka T, Sawatari H. Deep sedation predicts pressure injury in patients admitted to intensive care units. Nurs Crit Care 2022; 27:877-884. [PMID: 35048476 DOI: 10.1111/nicc.12753] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2021] [Revised: 11/22/2021] [Accepted: 01/04/2022] [Indexed: 02/03/2023]
Abstract
BACKGROUND Patients in intensive care units (ICU) are frequently prescribed sedatives, which might increase the risk for pressure injury (PI). Although the association between sedation and incidence of PI has been noted, the adequate sedation level to prevent the incidence of PI in patients admitted to ICU is still unclear. AIM This study aimed to investigate the association between fluctuating sedation levels and the incidence of PI in patients admitted to ICU. STUDY DESIGN We retrospectively reviewed the medical records of 104 patients admitted to ICU. Data regarding the length of ICU stay (LOS) and comorbid infection were abstracted from medical records. The Richmond Agitation-Sedation Scale (RASS) was scored twice per day, and the standardized RASS (S-RASS, summation of RASS values divided by the number of samples) was used to evaluate changes in sedation levels. RESULTS Among the 104 included patients, 65 patients (62.5%) were male (median age: 68.0 years), and 13 patients (12.5%) had PI during ICU admission. S-RASS scores were lower in patients with PI than in those without PI (P = .0001) even after adjustment for confounders (OR [95%CI]: 0.14 [0.03-0.58], P = .006). The LOS and infections were higher in patients with PI than in those without PI (P < .0001 and P = .005, respectively). The cut-off value of S-RASS for PI incidence was -3.2 (sensitivity: 88%; specificity: 85%), and a significant predictor of PI incidence (HR [95%CI]: 20.07 [2.53-159.11], P = .005). CONCLUSIONS Deeper sedation levels based on S-RASS scores, which account for the effects of fluctuating sedation levels, were a strong, highly accurate predictor of PI incidence in patients admitted to ICU. RELEVANCE TO CLINICAL PRACTICE Assessing fluctuations in the level of sedation using the S-RASS might help to identify sedative-induced PI in patients admitted to ICU.
Collapse
Affiliation(s)
- Yayoi Sasabe
- Department of Nursing, Hiroshima University Hospital, Hiroshima, Japan
| | - Mayumi Niitani
- Department of Nursing, Graduate School of Nursing, Yasuda Women's University, Hiroshima, Japan
| | - Chie Teramoto
- Department of Perioperative and Critical Care Management, Graduate School of Biomedical and Health Sciences, Hiroshima University, Hiroshima, Japan
| | - Satoshi Yamaga
- Department of Radiation Disaster Medicine, Research Institute for Radiation Biology and Medicine, Hiroshima University, Hiroshima, Japan
| | - Nobuaki Shime
- Department of Emergency and Critical Care Medicine, Graduate School of Biomedical Sciences, Hiroshima University, Hiroshima, Japan
| | - Kazuaki Tanabe
- Department of Perioperative and Critical Care Management, Graduate School of Biomedical and Health Sciences, Hiroshima University, Hiroshima, Japan
| | | | - Hiroyuki Sawatari
- Department of Perioperative and Critical Care Management, Graduate School of Biomedical and Health Sciences, Hiroshima University, Hiroshima, Japan
| |
Collapse
|
20
|
Torres-Soria AK, Romero Y, Balderas-Martínez YI, Velázquez-Cruz R, Torres-Espíndola LM, Camarena A, Flores-Soto E, Solís-Chagoyán H, Ruiz V, Carlos-Reyes Á, Salinas-Lara C, Luis-García ER, Chávez J, Castillejos-López M, Aquino-Gálvez A. Functional Repercussions of Hypoxia-Inducible Factor-2α in Idiopathic Pulmonary Fibrosis. Cells 2022; 11:cells11192938. [PMID: 36230900 PMCID: PMC9562026 DOI: 10.3390/cells11192938] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2022] [Revised: 09/06/2022] [Accepted: 09/07/2022] [Indexed: 11/16/2022] Open
Abstract
Hypoxia and hypoxia-inducible factors (HIFs) are essential in regulating several cellular processes, such as survival, differentiation, and the cell cycle; this adaptation is orchestrated in a complex way. In this review, we focused on the impact of hypoxia in the physiopathology of idiopathic pulmonary fibrosis (IPF) related to lung development, regeneration, and repair. There is robust evidence that the responses of HIF-1α and -2α differ; HIF-1α participates mainly in the acute phase of the response to hypoxia, and HIF-2α in the chronic phase. The analysis of their structure and of different studies showed a high specificity according to the tissue and the process involved. We propose that hypoxia-inducible transcription factor 2a (HIF-2α) is part of the persistent aberrant regeneration associated with developing IPF.
Collapse
Affiliation(s)
- Ana Karen Torres-Soria
- Red MEDICI, Carrera de Médico Cirujano, Facultad de Estudios Superiores de Iztacala Universidad Nacional Autónoma de México, Mexico City 54090, Mexico
| | - Yair Romero
- Facultad de Ciencias, Universidad Nacional Autónoma México, Mexico City 04510, Mexico
| | - Yalbi I. Balderas-Martínez
- Laboratorio de Biología Computacional, Instituto Nacional de Enfermedades Respiratorias Ismael Cosío Villegas, Mexico City 14080, Mexico
| | - Rafael Velázquez-Cruz
- Laboratorio de Genómica del Metabolismo Óseo, Instituto Nacional de Medicina Genómica, Mexico City 14610, Mexico
| | | | - Angel Camarena
- Laboratorio de HLA, Instituto Nacional de Enfermedades Respiratorias Ismael Cosío Villegas, Mexico City 04530, Mexico
| | - Edgar Flores-Soto
- Departamento de Farmacología, Facultad de Medicina, Universidad Nacional Autónoma de México, Mexico City 04510, Mexico
| | - Héctor Solís-Chagoyán
- Subdirección de Investigaciones Clínicas, Instituto Nacional de Psiquiatría Ramón de la Fuente Muñiz, Mexico City 14370, Mexico
| | - Víctor Ruiz
- Departamento de Fibrosis Pulmonar, Laboratorio de Biología Molecular, Instituto Nacional de Enfermedades Respiratorias Ismael Cosío Villegas, Mexico City 14080, Mexico
| | - Ángeles Carlos-Reyes
- Laboratorio de Onco-Inmunobiología, Departamento de Enfermedades Crónico-Degenerativas, Instituto Nacional de Enfermedades Respiratorias Ismael Cosío Villegas, Mexico City 14080, Mexico
| | - Citlaltepetl Salinas-Lara
- Red MEDICI, Carrera de Médico Cirujano, Facultad de Estudios Superiores de Iztacala Universidad Nacional Autónoma de México, Mexico City 54090, Mexico
| | - Erika Rubí Luis-García
- Departamento de Fibrosis Pulmonar, Laboratorio de Biología Celular, Instituto Nacional de Enfermedades Respiratorias Ismael Cosío Villegas, Mexico City 14080, Mexico
| | - Jaime Chávez
- Departamento de Hiperreactividad Bronquial, Instituto Nacional de Enfermedades, Respiratorias Ismael Cosío Villegas, Mexico City 14080, Mexico
| | - Manuel Castillejos-López
- Departamento de Epidemiología y Estadística, Instituto Nacional de Enfermedades, Respiratorias Ismael Cosío Villegas, Mexico City 14080, Mexico
- Correspondence: (M.C.-L.); (A.A.-G.)
| | - Arnoldo Aquino-Gálvez
- Departamento de Fibrosis Pulmonar, Laboratorio de Biología Molecular, Instituto Nacional de Enfermedades Respiratorias Ismael Cosío Villegas, Mexico City 14080, Mexico
- Correspondence: (M.C.-L.); (A.A.-G.)
| |
Collapse
|
21
|
Tsaur I, Thomas A, Monecke M, Zugelder M, Rutz J, Grein T, Maxeiner S, Xie H, Chun FKH, Rothweiler F, Cinatl J, Michaelis M, Haferkamp A, Blaheta RA. Amygdalin Exerts Antitumor Activity in Taxane-Resistant Prostate Cancer Cells. Cancers (Basel) 2022; 14:cancers14133111. [PMID: 35804883 PMCID: PMC9265127 DOI: 10.3390/cancers14133111] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2022] [Revised: 06/17/2022] [Accepted: 06/23/2022] [Indexed: 11/16/2022] Open
Abstract
Despite recent advances in the treatment of metastatic prostate cancer (PCa), resistance development after taxane treatments is inevitable, necessitating effective options to combat drug resistance. Previous studies indicated antitumoral properties of the natural compound amygdalin. However, whether amygdalin acts on drug-resistant tumor cells remains questionable. An in vitro study was performed to investigate the influence of amygdalin (10 mg/mL) on the growth of a panel of therapy-naïve and docetaxel- or cabazitaxel-resistant PCa cell lines (PC3, DU145, and LNCaP cells). Tumor growth, proliferation, clonal growth, and cell cycle progression were investigated. The cell cycle regulating proteins (phospho)cdk1, (phospho)cdk2, cyclin A, cyclin B, p21, and p27 and the mammalian target of rapamycin (mTOR) pathway proteins (phospho)Akt, (phospho)Raptor, and (phospho)Rictor as well as integrin β1 and the cytoskeletal proteins vimentin, ezrin, talin, and cytokeratin 8/18 were assessed. Furthermore, chemotactic activity and adhesion to extracellular matrix components were analyzed. Amygdalin dose-dependently inhibited tumor growth and reduced tumor clones in all (parental and resistant) PCa cell lines, accompanied by a G0/G1 phase accumulation. Cell cycle regulating proteins were significantly altered by amygdalin. A moderate influence of amygdalin on tumor cell adhesion and chemotaxis was observed as well, paralleled by modifications of cytoskeletal proteins and the integrin β1 expression level. Amygdalin may, therefore, block tumor growth and disseminative characteristics of taxane-resistant PCa cells. Further studies are warranted to determine amygdalin’s value as an antitumor drug.
Collapse
Affiliation(s)
- Igor Tsaur
- Department of Urology and Pediatric Urology, University Medicine Mainz, Langenbeckstr. 1, 55131 Mainz, Germany; (I.T.); (A.H.); (R.A.B.)
| | - Anita Thomas
- Department of Urology and Pediatric Urology, University Medicine Mainz, Langenbeckstr. 1, 55131 Mainz, Germany; (I.T.); (A.H.); (R.A.B.)
- Correspondence: ; Tel.: +49-6131-172312; Fax: +49-6131-173827
| | - Michelle Monecke
- Department of Urology, Goethe-University, 60590 Frankfurt am Main, Germany; (M.M.); (M.Z.); (J.R.); (T.G.); (S.M.); (H.X.); (F.K.-H.C.)
| | - Marion Zugelder
- Department of Urology, Goethe-University, 60590 Frankfurt am Main, Germany; (M.M.); (M.Z.); (J.R.); (T.G.); (S.M.); (H.X.); (F.K.-H.C.)
| | - Jochen Rutz
- Department of Urology, Goethe-University, 60590 Frankfurt am Main, Germany; (M.M.); (M.Z.); (J.R.); (T.G.); (S.M.); (H.X.); (F.K.-H.C.)
| | - Timothy Grein
- Department of Urology, Goethe-University, 60590 Frankfurt am Main, Germany; (M.M.); (M.Z.); (J.R.); (T.G.); (S.M.); (H.X.); (F.K.-H.C.)
| | - Sebastian Maxeiner
- Department of Urology, Goethe-University, 60590 Frankfurt am Main, Germany; (M.M.); (M.Z.); (J.R.); (T.G.); (S.M.); (H.X.); (F.K.-H.C.)
| | - Hui Xie
- Department of Urology, Goethe-University, 60590 Frankfurt am Main, Germany; (M.M.); (M.Z.); (J.R.); (T.G.); (S.M.); (H.X.); (F.K.-H.C.)
| | - Felix K.-H. Chun
- Department of Urology, Goethe-University, 60590 Frankfurt am Main, Germany; (M.M.); (M.Z.); (J.R.); (T.G.); (S.M.); (H.X.); (F.K.-H.C.)
| | - Florian Rothweiler
- Institute of Medical Virology, Goethe-University, 60596 Frankfurt am Main, Germany; (F.R.)
- Petra Joh-Forschungshaus, 60528 Frankfurt am Main, Germany
| | - Jindrich Cinatl
- Institute of Medical Virology, Goethe-University, 60596 Frankfurt am Main, Germany; (F.R.)
- Petra Joh-Forschungshaus, 60528 Frankfurt am Main, Germany
| | - Martin Michaelis
- Industrial Biotechnology Centre, School of Biosciences, University of Kent, Canterbury CT2 7NJ, UK;
| | - Axel Haferkamp
- Department of Urology and Pediatric Urology, University Medicine Mainz, Langenbeckstr. 1, 55131 Mainz, Germany; (I.T.); (A.H.); (R.A.B.)
| | - Roman A. Blaheta
- Department of Urology and Pediatric Urology, University Medicine Mainz, Langenbeckstr. 1, 55131 Mainz, Germany; (I.T.); (A.H.); (R.A.B.)
| |
Collapse
|
22
|
CTK7A, a curcumin derivative, can be a potential candidate for targeting HIF-1α/p300 complex: Evidences from in vitro and computational studies. Biophys Chem 2022; 287:106828. [DOI: 10.1016/j.bpc.2022.106828] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2022] [Revised: 05/07/2022] [Accepted: 05/13/2022] [Indexed: 11/18/2022]
|
23
|
Dimethyloxalylglycine (DMOG), a Hypoxia Mimetic Agent, Does Not Replicate a Rat Pheochromocytoma (PC12) Cell Biological Response to Reduced Oxygen Culture. Biomolecules 2022; 12:biom12040541. [PMID: 35454130 PMCID: PMC9027160 DOI: 10.3390/biom12040541] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2021] [Revised: 03/25/2022] [Accepted: 03/30/2022] [Indexed: 02/01/2023] Open
Abstract
Cells respond to reduced oxygen availability predominately by activation of the hypoxia-inducible factor (HIF) pathway. HIF activation upregulates hundreds of genes that help cells survive in the reduced oxygen environment. The aim of this study is to determine whether chemical-induced HIF accumulation mimics all aspects of the hypoxic response of cells. We compared the effects of dimethyloxalylglycine (DMOG) (a HIF stabiliser) on PC12 cells cultured in air oxygen (20.9% O2, AO) with those cultured in either intermittent 20.9% O2 to 2% O2 (IH) or constant 2% O2 (CN). Cell viability, cell cycle, HIF accumulation, reactive oxygen species (ROS) formation, mitochondrial function and differentiation were used to characterise the PC12 cells and evaluate the impact of DMOG. IH and CN culture reduced the increase in cell numbers after 72 and 96 h and MTT activity after 48 h compared to AO culture. Further, DMOG supplementation in AO induced a dose-dependent reduction in the increase in PC12 cell numbers and MTT activity. IH-cultured PC12 cells displayed increased and sustained HIF-1 expression over 96 h. This was accompanied by increased ROS and mitochondrial burden. PC12 cells in CN displayed little changes in HIF-1 expression or ROS levels. DMOG (0.1 mM) supplementation resulted in an IH-like HIF-1 profile. The mitochondrial burden and action potential of DMOG-supplemented PC12 cells did not mirror those seen in other conditions. DMOG significantly increased S phase cell populations after 72 and 96 h. No significant effect on PC12 cell differentiation was noted with IH and CN culture without induction by nerve growth factor (NGF), while DMOG significantly increased PC12 cell differentiation with and without NGF. In conclusion, DMOG and reduced oxygen levels stabilise HIF and affect mitochondrial activity and cell behaviour. However, DMOG does not provide an accurate replication of the reduced oxygen environments.
Collapse
|
24
|
Hashimoto T, Urushihara Y, Murata Y, Fujishima Y, Hosoi Y. AMPK increases expression of ATM through transcriptional factor Sp1 and induces radioresistance under severe hypoxia in glioblastoma cell lines. Biochem Biophys Res Commun 2022; 590:82-88. [PMID: 34973534 DOI: 10.1016/j.bbrc.2021.12.076] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2021] [Accepted: 12/21/2021] [Indexed: 11/24/2022]
Abstract
We have previously reported that severe hypoxia increases expression and activity of the DNA damage sensor ATM by activation of the key energy sensor AMPK. Here, to elucidate molecular mechanisms underlying increased expression and activity of ATM by AMPK under severe hypoxia, we investigated roles of transcriptional factors Sp1 and FoxO3a using human glioblastoma cell lines T98G and A172. Severe hypoxia increased expression of ATM, AMPKα and Sp1 but not that of FoxO3a. Knockdown of AMPKα suppressed expression of ATM and Sp1 and suppressed cellular radioresistance under severe hypoxia without affecting cell cycle distribution. Knockdown of Sp1 suppressed expression of ATM. These results suggest that increased expression and activity of AMPK under severe hypoxia induce cellular radioresistance through AMPK/Sp1/ATM pathway.
Collapse
Affiliation(s)
- Takuma Hashimoto
- Department of Radiation Biology, Tohoku University School of Medicine, 2-1 Seiryo-machi, Aoba-ku, Sendai, Miyagi, 980-8575, Japan
| | - Yusuke Urushihara
- Department of Radiation Biology, Tohoku University School of Medicine, 2-1 Seiryo-machi, Aoba-ku, Sendai, Miyagi, 980-8575, Japan
| | - Yasuhiko Murata
- MSD K.K., 1-13-12 Kudankita, Chiyoda-ku, Tokyo, 102-8667, Japan
| | - Yohei Fujishima
- Department of Risk Analysis and Biodosimetry, Institute of Radiation Emergency Medicine, Hirosaki University, 66-1 Hon-cho, Hirosaki-shi, Aomori, 036-8564, Japan
| | - Yoshio Hosoi
- Department of Radiation Biology, Tohoku University School of Medicine, 2-1 Seiryo-machi, Aoba-ku, Sendai, Miyagi, 980-8575, Japan.
| |
Collapse
|
25
|
Kim Y, Kim S, Im G, Kim YH, Jeong G, Jeon HR, Kim D, Lee H, Park SY, Cho SM, Bhang SH. Area light source-triggered latent angiogenic molecular mechanisms intensify therapeutic efficacy of adult stem cells. Bioeng Transl Med 2022; 7:e10255. [PMID: 35079630 PMCID: PMC8780080 DOI: 10.1002/btm2.10255] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2021] [Revised: 09/03/2021] [Accepted: 09/05/2021] [Indexed: 12/31/2022] Open
Abstract
Light-based therapy such as photobiomodulation (PBM) reportedly produces beneficial physiological effects in cells and tissues. However, most reports have focused on the immediate and instant effects of light. Considering the physiological effects of natural light exposure in living organisms, the latent reaction period after irradiation should be deliberated. In contrast to previous reports, we examined the latent reaction period after light exposure with optimized irradiating parameters and validated novel therapeutic molecular mechanisms for the first time. we demonstrated an organic light-emitting diode (OLED)-based PBM (OPBM) strategy that enhances the angiogenic efficacy of human adipose-derived stem cells (hADSCs) via direct irradiation with red OLEDs of optimized wavelength, voltage, current, luminance, and duration, and investigated the underlying molecular mechanisms. Our results revealed that the angiogenic paracrine effect, viability, and adhesion of hADSCs were significantly intensified by our OPBM strategy. Following OPBM treatment, significant changes were observed in HIF-1α expression, intracellular reactive oxygen species levels, activation of the receptor tyrosine kinase, and glycolytic pathways in hADSCs. In addition, transplantation of OLED-irradiated hADSCs resulted in significantly enhanced limb salvage ratio in a mouse model of hindlimb ischemia. Our OPBM might serve as a new paradigm for stem cell culture systems to develop cell-based therapies in the future.
Collapse
Affiliation(s)
- Yu‐Jin Kim
- School of Chemical EngineeringSungkyunkwan UniversitySuwonRepublic of Korea
| | - Sung‐Won Kim
- School of Chemical EngineeringSungkyunkwan UniversitySuwonRepublic of Korea
| | - Gwang‐Bum Im
- School of Chemical EngineeringSungkyunkwan UniversitySuwonRepublic of Korea
| | - Yeong Hwan Kim
- School of Chemical EngineeringSungkyunkwan UniversitySuwonRepublic of Korea
| | - Gun‐Jae Jeong
- Division of Vascular Surgery, Samsung Medical CenterSungkyunkwan University School of MedicineSeoulRepublic of Korea
| | - Hye Ran Jeon
- Department of Health Sciences and Technology, SAIHSTSungkyunkwan UniversitySeoulRepublic of Korea
| | - Dong‐Ik Kim
- Division of Vascular Surgery, Samsung Medical CenterSungkyunkwan University School of MedicineSeoulRepublic of Korea
| | - Haeshin Lee
- Department of Chemistry, Center for Nature‐Inspired Technology (CNiT)Korea Advanced Institute of Science and Technology (KAIST)DaejeonRepublic of Korea
| | - Sung Young Park
- Department of Chemical and Biological EngineeringKorea National University of TransportationChungjuRepublic of Korea
| | - Sung Min Cho
- School of Chemical EngineeringSungkyunkwan UniversitySuwonRepublic of Korea
| | - Suk Ho Bhang
- School of Chemical EngineeringSungkyunkwan UniversitySuwonRepublic of Korea
| |
Collapse
|
26
|
de Keijzer MJ, de Klerk DJ, de Haan LR, van Kooten RT, Franchi LP, Dias LM, Kleijn TG, van Doorn DJ, Heger M. Inhibition of the HIF-1 Survival Pathway as a Strategy to Augment Photodynamic Therapy Efficacy. Methods Mol Biol 2022; 2451:285-403. [PMID: 35505024 DOI: 10.1007/978-1-0716-2099-1_19] [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: 06/14/2023]
Abstract
Photodynamic therapy (PDT) is a non-to-minimally invasive treatment modality that utilizes photoactivatable drugs called photosensitizers to disrupt tumors with locally photoproduced reactive oxygen species (ROS). Photosensitizer activation by light results in hyperoxidative stress and subsequent tumor cell death, vascular shutdown and hypoxia, and an antitumor immune response. However, sublethally afflicted tumor cells initiate several survival mechanisms that account for decreased PDT efficacy. The hypoxia inducible factor 1 (HIF-1) pathway is one of the most effective cell survival pathways that contributes to cell recovery from PDT-induced damage. Several hundred target genes of the HIF-1 heterodimeric complex collectively mediate processes that are involved in tumor cell survival directly and indirectly (e.g., vascularization, glucose metabolism, proliferation, and metastasis). The broad spectrum of biological ramifications culminating from the activation of HIF-1 target genes reflects the importance of HIF-1 in the context of therapeutic recalcitrance. This chapter elaborates on the involvement of HIF-1 in cancer biology, the hypoxic response mechanisms, and the role of HIF-1 in PDT. An overview of inhibitors that either directly or indirectly impede HIF-1-mediated survival signaling is provided. The inhibitors may be used as pharmacological adjuvants in combination with PDT to augment therapeutic efficacy.
Collapse
Affiliation(s)
- Mark J de Keijzer
- Jiaxing Key Laboratory for Photonanomedicine and Experimental Therapeutics, Department of Pharmaceutics, College of Medicine, Jiaxing University, Jiaxing, Zhejiang, People's Republic of China
- Department of Pharmaceutics, Utrecht Institute of Pharmaceutical Sciences, Utrecht University, Utrecht, The Netherlands
| | - Daniel J de Klerk
- Jiaxing Key Laboratory for Photonanomedicine and Experimental Therapeutics, Department of Pharmaceutics, College of Medicine, Jiaxing University, Jiaxing, Zhejiang, People's Republic of China
- Laboratory of Experimental Oncology, Department of Pathology, Erasmus MC, Rotterdam, The Netherlands
| | - Lianne R de Haan
- Laboratory of Experimental Oncology, Department of Pathology, Erasmus MC, Rotterdam, The Netherlands
| | - Robert T van Kooten
- Department of Surgery, Leiden University Medical Center, Leiden, The Netherlands
| | - Leonardo P Franchi
- Departamento de Bioquímica e Biologia Molecular, Instituto de Ciências Biológicas (ICB) 2, Universidade Federal de Goiás (UFG), Goiânia, GO, Brazil
- Faculty of Philosophy, Sciences, and Letters of Ribeirão Preto, epartment of Chemistry, Center of Nanotechnology and Tissue Engineering-Photobiology and Photomedicine Research Group,University of São Paulo, São Paulo, Brazil
| | - Lionel M Dias
- Jiaxing Key Laboratory for Photonanomedicine and Experimental Therapeutics, Department of Pharmaceutics, College of Medicine, Jiaxing University, Jiaxing, Zhejiang, People's Republic of China
- Laboratory of Experimental Oncology, Department of Pathology, Erasmus MC, Rotterdam, The Netherlands
| | - Tony G Kleijn
- Jiaxing Key Laboratory for Photonanomedicine and Experimental Therapeutics, Department of Pharmaceutics, College of Medicine, Jiaxing University, Jiaxing, Zhejiang, People's Republic of China
- Laboratory of Experimental Oncology, Department of Pathology, Erasmus MC, Rotterdam, The Netherlands
| | - Diederick J van Doorn
- Department of Gastroenterology and Hepatology, Amsterdam UMC, Location AMC, University of Amsterdam, Amsterdam, The Netherlands
| | - Michal Heger
- Jiaxing Key Laboratory for Photonanomedicine and Experimental Therapeutics, Department of Pharmaceutics, College of Medicine, Jiaxing University, Jiaxing, Zhejiang, People's Republic of China.
- Department of Pharmaceutics, Utrecht Institute of Pharmaceutical Sciences, Utrecht University, Utrecht, The Netherlands.
- Laboratory of Experimental Oncology, Department of Pathology, Erasmus MC, Rotterdam, The Netherlands.
| |
Collapse
|
27
|
Lu D, Yang N, Wang S, Liu W, Zhang D, Wang J, Huang B, Li X. Identifying the Predictive Role of Oxidative Stress Genes in the Prognosis of Glioma Patients. Med Sci Monit 2021; 27:e934161. [PMID: 34836934 PMCID: PMC8634738 DOI: 10.12659/msm.934161] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022] Open
Abstract
Background Gliomas are primary aggressive brain tumors with poor prognoses. Oxidative stress plays a crucial role in the tumorigenesis and drug resistance of gliomas. The aim of the present study was to use integrated bioinformatics analyses to evaluate the prognostic value of oxidative stress-related genes (OSRGs) in glioma. Material/Methods Disease- and prognosis-associated OSRGs were identified using microarray and clinical data from the Chinese Glioma Genome Atlas database. Functional enrichment, gene-gene interaction, protein-protein interaction, and survival analyses were performed in screened OSRGs. The protein expression was validated by the Human Protein Atlas database. A risk score model was constructed and verified through Cox regression, receiver operating characteristic curve, principal component, and stratified analyses. The Cancer Genome Atlas (TCGA) database was used for external validation. A nomogram was constructed to facilitate the clinical application. Results Twenty-one disease-associated and 14 prognosis-associated OSRGs were identified. Enrichment analyses indicated that these signature OSRGs were involved in tumorigenesis and drug resistance of glioma. The risk score model demonstrated a significant difference in overall survival between the high- and low-risk groups. The area under the curve and hazard ratio (1.296) revealed the independent prognostic value of the model. The model exhibited good predictive efficacy in the TCGA cohort. A clinical nomogram was constructed to calculate survival rates in glioma patients at 1, 3, and 5 years. Conclusions Our comprehensive study indicated that OSRGs were valuable for prognosis prediction in glioma, which provides a novel insight into the relationship between oxidative stress and glioma and a potential therapeutic strategy for glioma patients.
Collapse
Affiliation(s)
- Di Lu
- Department of Neurosurgery, Qilu Hospital, Cheeloo College of Medicine, Shandong University and Institute of Brain and Brain-Inspired Science, Shandong University, Jinan, Shandong, China (mainland).,Key Laboratory of Brain Function Remodeling, Qilu Hospital of Shandong University, Jinan, Shandong, China (mainland)
| | - Ning Yang
- Department of Neurosurgery, Qilu Hospital, Cheeloo College of Medicine, Shandong University and Institute of Brain and Brain-Inspired Science, Shandong University, Jinan, Shandong, China (mainland).,Key Laboratory of Brain Function Remodeling, Qilu Hospital of Shandong University, Jinan, Shandong, China (mainland)
| | - Shuai Wang
- Department of Neurosurgery, Qilu Hospital, Cheeloo College of Medicine, Shandong University and Institute of Brain and Brain-Inspired Science, Shandong University, Jinan, Shandong, China (mainland).,Key Laboratory of Brain Function Remodeling, Qilu Hospital of Shandong University, Jinan, Shandong, China (mainland)
| | - Wenyu Liu
- Department of Neurosurgery, Qilu Hospital, Cheeloo College of Medicine, Shandong University and Institute of Brain and Brain-Inspired Science, Shandong University, Jinan, Shandong, China (mainland).,Key Laboratory of Brain Function Remodeling, Qilu Hospital of Shandong University, Jinan, Shandong, China (mainland)
| | - Di Zhang
- Department of Neurosurgery, Qilu Hospital, Cheeloo College of Medicine, Shandong University and Institute of Brain and Brain-Inspired Science, Shandong University, Jinan, Shandong, China (mainland).,Key Laboratory of Brain Function Remodeling, Qilu Hospital of Shandong University, Jinan, Shandong, China (mainland)
| | - Jian Wang
- Department of Neurosurgery, Qilu Hospital, Cheeloo College of Medicine, Shandong University and Institute of Brain and Brain-Inspired Science, Shandong University, Jinan, Shandong, China (mainland).,Key Laboratory of Brain Function Remodeling, Qilu Hospital of Shandong University, Jinan, Shandong, China (mainland).,Department of Biomedicine, University of Bergen, Bergen, Norway
| | - Bin Huang
- Department of Neurosurgery, Qilu Hospital, Cheeloo College of Medicine, Shandong University and Institute of Brain and Brain-Inspired Science, Shandong University, Jinan, Shandong, China (mainland).,Key Laboratory of Brain Function Remodeling, Qilu Hospital of Shandong University, Jinan, Shandong, China (mainland)
| | - Xingang Li
- Department of Neurosurgery, Qilu Hospital, Cheeloo College of Medicine, Shandong University and Institute of Brain and Brain-Inspired Science, Shandong University, Jinan, Shandong, China (mainland).,Key Laboratory of Brain Function Remodeling, Qilu Hospital of Shandong University, Jinan, Shandong, China (mainland)
| |
Collapse
|
28
|
Le N, Hufford TM, Park JS, Brewster RM. Differential expression and hypoxia-mediated regulation of the N-myc downstream regulated gene family. FASEB J 2021; 35:e21961. [PMID: 34665878 PMCID: PMC8573611 DOI: 10.1096/fj.202100443r] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2021] [Revised: 09/11/2021] [Accepted: 09/15/2021] [Indexed: 01/09/2023]
Abstract
Many organisms rely on oxygen to generate cellular energy (adenosine triphosphate or ATP). During severe hypoxia, the production of ATP decreases, leading to cell damage or death. Conversely, excessive oxygen causes oxidative stress that is equally damaging to cells. To mitigate pathological outcomes, organisms have evolved mechanisms to adapt to fluctuations in oxygen levels. Zebrafish embryos are remarkably hypoxia-tolerant, surviving anoxia (zero oxygen) for hours in a hypometabolic, energy-conserving state. To begin to unravel underlying mechanisms, we analyze here the distribution of the N-myc Downstream Regulated Gene (ndrg) family, ndrg1-4, and their transcriptional response to hypoxia. These genes have been primarily studied in cancer cells and hence little is understood about their normal function and regulation. We show here using in situ hybridization that ndrgs are expressed in metabolically demanding organs of the zebrafish embryo, such as the brain, kidney, and heart. To investigate whether ndrgs are hypoxia-responsive, we exposed embryos to different durations and severity of hypoxia and analyzed transcript levels. We observed that ndrgs are differentially regulated by hypoxia and that ndrg1a has the most robust response, with a ninefold increase following prolonged anoxia. We further show that this treatment resulted in de novo expression of ndrg1a in tissues where the transcript is not observed under normoxic conditions and changes in Ndrg1a protein expression post-reoxygenation. These findings provide an entry point into understanding the role of this conserved gene family in the adaptation of normal cells to hypoxia and reoxygenation.
Collapse
Affiliation(s)
- Nguyet Le
- Department of Biological SciencesUniversity of Maryland, Baltimore CountyBaltimoreMarylandUSA
| | - Timothy M. Hufford
- Department of Biological SciencesUniversity of Maryland, Baltimore CountyBaltimoreMarylandUSA
| | - Jong S. Park
- Department of Biological SciencesUniversity of Maryland, Baltimore CountyBaltimoreMarylandUSA
| | - Rachel M. Brewster
- Department of Biological SciencesUniversity of Maryland, Baltimore CountyBaltimoreMarylandUSA
| |
Collapse
|
29
|
Ross SH, Rollings CM, Cantrell DA. Quantitative Analyses Reveal How Hypoxia Reconfigures the Proteome of Primary Cytotoxic T Lymphocytes. Front Immunol 2021; 12:712402. [PMID: 34603285 PMCID: PMC8484760 DOI: 10.3389/fimmu.2021.712402] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2021] [Accepted: 08/26/2021] [Indexed: 12/18/2022] Open
Abstract
Metabolic and nutrient-sensing pathways play an important role in controlling the efficacy of effector T cells. Oxygen is a critical regulator of cellular metabolism. However, during immune responses T cells must function in oxygen-deficient, or hypoxic, environments. Here, we used high resolution mass spectrometry to investigate how the proteome of primary murine CD8+ cytotoxic T lymphocytes (CTLs) is reconfigured in response to hypoxia in vitro. We identified and quantified over 7,600 proteins and discovered that hypoxia increased the abundance of a selected number of proteins in CTLs. This included glucose transporters, metabolic enzymes, transcription factors, cytolytic effector molecules, checkpoint receptors and adhesion molecules. While some of these proteins may augment the effector functions of CTLs, others may limit their cytotoxicity. Moreover, we determined that hypoxia could inhibit IL-2-induced proliferation cues and antigen-induced pro-inflammatory cytokine production in CTLs. These data provide a comprehensive resource for understanding the magnitude of the CTL response to hypoxia and emphasise the importance of oxygen-sensing pathways for controlling CD8+ T cells. Additionally, this study provides new understanding about how hypoxia may promote the effector function of CTLs, while contributing to their dysfunction in some contexts.
Collapse
Affiliation(s)
- Sarah H Ross
- Immunology Programme, The Babraham Institute, Cambridge, United Kingdom.,Division of Cell Signalling and Immunology, School of Life Sciences, University of Dundee, Dundee, United Kingdom
| | - Christina M Rollings
- Division of Cell Signalling and Immunology, School of Life Sciences, University of Dundee, Dundee, United Kingdom
| | - Doreen A Cantrell
- Division of Cell Signalling and Immunology, School of Life Sciences, University of Dundee, Dundee, United Kingdom
| |
Collapse
|
30
|
Ivanova JS, Pugovkina NA, Neganova IE, Kozhukharova IV, Nikolsky NN, Lyublinskaya OG. Cell cycle-coupled changes in the level of reactive oxygen species support the proliferation of human pluripotent stem cells. Stem Cells 2021; 39:1671-1687. [PMID: 34460135 DOI: 10.1002/stem.3450] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2021] [Accepted: 08/10/2021] [Indexed: 12/12/2022]
Abstract
The study of proliferation regulation in human pluripotent stem cells is crucial to gain insights into understanding the physiology of these cells. However, redox regulation of the pluripotent cell cycle remains largely unexplored. Here, using human embryonic stem cells (hESCs) as well as human induced pluripotent stem cells (hiPSCs), we demonstrate that the level of reactive oxygen species (ROS) in pluripotent cells oscillates in accordance with the cell cycle progression with the peak occurring at transition from S to G2 /M phase of the cycle. A decrease of this level by antioxidants leads to hindered S-phase initiation and progression but does not affect the early-G1 -phase or mitosis. Cells exposed to antioxidants in the early-G1 -phase accumulate the phosphorylated retinoblastoma protein and overcome the restriction point but are unable to accumulate the main regulators of the S phase-CYCLIN A and GEMININ. Based on the previous findings that CYCLIN A stability is affected by redox homeostasis disturbances in somatic cells, we compared the responses to antioxidant treatments in hESCs and in their differentiated fibroblast-like progeny cells (difESCs). In difESCs, similar to hESCs, a decrease in ROS level results in the disruption of S-phase initiation accompanied by a deficiency of the CYCLIN A level. Moreover, in antioxidant-treated cells, we revealed the accumulation of DNA breaks, which was accompanied by activation of the apoptosis program in pluripotent cells. Thus, we conclude that maintaining the physiological ROS level is essential for promotion of proliferation and accurate DNA synthesis in pluripotent cells and their differentiated descendants.
Collapse
Affiliation(s)
- Julia S Ivanova
- Institute of Cytology of the Russian Academy of Sciences, St. Petersburg, Russia
| | - Natalia A Pugovkina
- Institute of Cytology of the Russian Academy of Sciences, St. Petersburg, Russia
| | - Irina E Neganova
- Institute of Cytology of the Russian Academy of Sciences, St. Petersburg, Russia
| | - Irina V Kozhukharova
- Institute of Cytology of the Russian Academy of Sciences, St. Petersburg, Russia
| | - Nikolay N Nikolsky
- Institute of Cytology of the Russian Academy of Sciences, St. Petersburg, Russia
| | - Olga G Lyublinskaya
- Institute of Cytology of the Russian Academy of Sciences, St. Petersburg, Russia
| |
Collapse
|
31
|
Li C, Liu Z, Wu G, Zang Z, Zhang JQ, Li X, Tao J, Shen M, Liu H. FOXO1 mediates hypoxia-induced G0/G1 arrest in ovarian somatic granulosa cells by activating the TP53INP1-p53-CDKN1A pathway. Development 2021; 148:269228. [PMID: 34152408 DOI: 10.1242/dev.199453] [Citation(s) in RCA: 21] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2021] [Accepted: 06/13/2021] [Indexed: 12/14/2022]
Abstract
The development of ovarian follicles constitutes the foundation of female reproduction. The proliferation of granulosa cells (GCs) is a basic process required to ensure normal follicular development. However, the mechanisms involved in controlling GC cell cycle are not fully understood. Here, by performing gene expression profiling in the domestic pig (Sus scrofa), we showed that cell cycle arrest at G0/G1 phase is highly correlated with pathways associated with hypoxic stress and FOXO signalling. Specifically, the elevated proportion of GCs at the arrested G0/G1 phase was accompanied by increased nuclear translocation of FOXO1 under conditions of hypoxia both in vivo and in vitro. Furthermore, phosphorylation of 14-3-3 by the JNK kinase is required for hypoxia-mediated FOXO1 activation and the resultant G0/G1 arrest. Notably, a FOXO1 mutant without DNA-binding activity failed to induce G0/G1 arrest of GCs during hypoxia. Importantly, we identified a new target gene of FOXO1, namely TP53INP1, which contributes to suppression of the G1-S cell cycle transition in response to hypoxia. Furthermore, we demonstrated that the inhibitory effect of the FOXO1-TP53INP1 axis on the GC cell cycle is mediated through a p53-CDKN1A-dependent mechanism. These findings could provide avenues for the clinical treatment of human infertility caused by impaired follicular development.
Collapse
Affiliation(s)
- Chengyu Li
- College of Animal Science and Technology, Nanjing Agricultural University, Nanjing 210095, China
| | - Zhaojun Liu
- College of Animal Science and Technology, Nanjing Agricultural University, Nanjing 210095, China
| | - Gang Wu
- College of Animal Science and Technology, Nanjing Agricultural University, Nanjing 210095, China
| | - Ziyu Zang
- College of Animal Science and Technology, Nanjing Agricultural University, Nanjing 210095, China
| | - Jia-Qing Zhang
- Institute of Animal Husbandry and Veterinary Science, Henan Academy of Agricultural Sciences, Zhengzhou, 450002, China
| | - Xiaoxuan Li
- College of Animal Science and Technology, Nanjing Agricultural University, Nanjing 210095, China
| | - Jingli Tao
- College of Animal Science and Technology, Nanjing Agricultural University, Nanjing 210095, China
| | - Ming Shen
- College of Animal Science and Technology, Nanjing Agricultural University, Nanjing 210095, China
| | - Honglin Liu
- College of Animal Science and Technology, Nanjing Agricultural University, Nanjing 210095, China
| |
Collapse
|
32
|
Interplay between Hypoxia and Extracellular Vesicles in Cancer and Inflammation. BIOLOGY 2021; 10:biology10070606. [PMID: 34209290 PMCID: PMC8301089 DOI: 10.3390/biology10070606] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/31/2021] [Revised: 06/24/2021] [Accepted: 06/24/2021] [Indexed: 12/13/2022]
Abstract
Simple Summary Mounting evidence suggests a role for extracellular vesicles in cell-to-cell communication, in both physiological and pathological conditions. Moreover, the molecular content of vesicles can be exploited for diagnostic and therapeutic purposes. Inflamed tissues and tumors are often characterized by hypoxic areas, where oxygen levels drop dramatically. Several studies demonstrated that hypoxic stress affects the release of vesicles and their content. This review is intended to provide an exhaustive overview on the relationship between hypoxia and vesicles in inflammatory diseases and cancer. Abstract Hypoxia is a severe stress condition often observed in cancer and chronically inflamed cells and tissues. Extracellular vesicles play pivotal roles in these pathological processes and carry biomolecules that can be detected in many biofluids and may be exploited for diagnostic purposes. Several studies report the effects of hypoxia on extracellular vesicles’ release, molecular content, and biological functions in disease. This review summarizes the most recent findings in this field, highlighting the areas that warrant further investigation.
Collapse
|
33
|
Li C, Liu Z, Zhou J, Meng X, Liu S, Li W, Zhang X, Zhou J, Yao W, Dong C, Cao Y, Li R, Chen B, Jiang A, Jiang Y, Ning C, Zhao F, Wei Y, Sun SC, Tao J, Wu W, Shen M, Liu H. Insulin-like growth factor-I prevents hypoxia-inducible factor-1 alpha-dependent G1/S arrest by activating cyclin E/cyclin-dependent kinase2 via the phoshatidylinositol-3 kinase/AKT/forkhead box O1/Cdkn1b pathway in porcine granulosa cells†. Biol Reprod 2021; 102:116-132. [PMID: 31435642 DOI: 10.1093/biolre/ioz162] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2019] [Revised: 05/23/2019] [Accepted: 08/12/2019] [Indexed: 11/13/2022] Open
Abstract
As the follicle develops, the thickening of the granulosa compartment leads to progressively deficient supply of oxygen in granulosa cells (GCs) due to the growing distances from the follicular vessels. These conditions are believed to cause hypoxia in GCs during folliculogenesis. Upon hypoxic conditions, several types of mammalian cells have been reported to undergo cell cycle arrest. However, it remains unclear whether hypoxia exerts any impact on cell cycle progression of GCs. On the other hand, although the GCs may live in a hypoxic environment, their mitotic capability appears to be unaffected in growing follicles. It thus raises the question whether there are certain intraovarian factors that might overcome the inhibitory effects of hypoxia. The present study provides the first evidence suggesting that cobalt chloride (CoCl2)-mimicked hypoxia prevented G1-to-S cell cycle progression in porcine GCs. In addition, we demonstrated that the inhibitory effects of CoCl2 on GCs cell cycle are mediated through hypoxia-inducible factor-1 alpha/FOXO1/Cdkn1b pathway. Moreover, we identified insulin-like growth factor-I (IGF-I) as an intrafollicular factor required for cell cycle recovery by binding to IGF-I receptor in GCs suffering CoCl2 stimulation. Further investigations confirmed a role of IGF-I in preserving G1/S progression of CoCl2-treated GCs via activating the cyclin E/cyclin-dependent kinase2 complex through the phoshatidylinositol-3 kinase/protein kinase B (AKT)/FOXO1/Cdkn1b axis. Although the present findings were based on a hypoxia mimicking model by using CoCl2, our study might shed new light on the regulatory mechanism of GCs cell cycle upon hypoxic stimulation.
Collapse
Affiliation(s)
- Chengyu Li
- College of Animal Science and Technology, Nanjing Agricultural University, Nanjing 210095, China
| | - Zhaojun Liu
- College of Animal Science and Technology, Nanjing Agricultural University, Nanjing 210095, China
| | - Jiaqi Zhou
- College of Animal Science and Technology, Nanjing Agricultural University, Nanjing 210095, China
| | - Xueqin Meng
- College of Animal Science and Technology, Nanjing Agricultural University, Nanjing 210095, China
| | - Shuo Liu
- College of Animal Science and Technology, Nanjing Agricultural University, Nanjing 210095, China
| | - Weijian Li
- College of Animal Science and Technology, Nanjing Agricultural University, Nanjing 210095, China
| | - Xue Zhang
- College of Animal Science and Technology, Nanjing Agricultural University, Nanjing 210095, China
| | - Jilong Zhou
- Institute of Stem Cell and Regenerative Biology, College of Animal Science and Veterinary Medicine, Huazhong Agricultural University, Wuhan 430070, China
| | - Wang Yao
- College of Animal Science and Technology, Nanjing Agricultural University, Nanjing 210095, China
| | - Chao Dong
- College of Animal Science and Technology, Nanjing Agricultural University, Nanjing 210095, China
| | - Yan Cao
- College of Animal Science and Technology, Nanjing Agricultural University, Nanjing 210095, China
| | - Rongyang Li
- College of Animal Science and Technology, Nanjing Agricultural University, Nanjing 210095, China
| | - Baobao Chen
- College of Animal Science and Technology, Nanjing Agricultural University, Nanjing 210095, China
| | - Aiwen Jiang
- College of Animal Science and Technology, Nanjing Agricultural University, Nanjing 210095, China
| | - Yi Jiang
- College of Animal Science and Technology, Nanjing Agricultural University, Nanjing 210095, China
| | - Caibo Ning
- College of Animal Science and Technology, Nanjing Agricultural University, Nanjing 210095, China
| | - Fang Zhao
- College of Animal Science and Technology, Nanjing Agricultural University, Nanjing 210095, China
| | - Yinghui Wei
- College of Animal Science and Technology, Nanjing Agricultural University, Nanjing 210095, China
| | - Shao-Chen Sun
- College of Animal Science and Technology, Nanjing Agricultural University, Nanjing 210095, China
| | - Jingli Tao
- College of Animal Science and Technology, Nanjing Agricultural University, Nanjing 210095, China
| | - Wangjun Wu
- College of Animal Science and Technology, Nanjing Agricultural University, Nanjing 210095, China
| | - Ming Shen
- College of Animal Science and Technology, Nanjing Agricultural University, Nanjing 210095, China
| | - Honglin Liu
- College of Animal Science and Technology, Nanjing Agricultural University, Nanjing 210095, China
| |
Collapse
|
34
|
Immunotherapy Using Oxygenated Water and Tumor-Derived Exosomes Potentiates Antitumor Immune Response and Attenuates Malignancy Tendency in Mice Model of Breast Cancer. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2021; 2021:5529484. [PMID: 34194604 PMCID: PMC8181112 DOI: 10.1155/2021/5529484] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 02/26/2021] [Revised: 03/22/2021] [Accepted: 04/23/2021] [Indexed: 01/16/2023]
Abstract
Breast cancer is one of the most common type of tumor and the leading cause of death in the world's female population. Various therapeutic approaches have been used to treat tumors but have not led to complete recovery and have even damaged normal cells in the body. Moreover, metastatic tumors such as breast cancer are much more resistant to treatment, and current treatments have not been very successful in treating them and remain a challenge. Therefore, new approaches should be applied to overcome this problem. Given the importance of hypoxia in tumor survival, we aimed to test the antitumor effects of oxygenated water to decrease hypoxia along with tumor-derived exosomes to target tumor. The purpose of administering oxygenated water and tumor exosomes was to reduce hypoxia and establish an effective immune response against tumor antigens, respectively. For this purpose, the breast cancer mice model was induced using the 4T1 cell line in Balb/c mice and treated with oxygenated water via an intratumoral (IT) and/or intraperitoneal (IP) route and/or exosome (TEX). Oxygenation via the IT+IP route was more efficient than oxygenation via the IT or IP route. The efficiency of oxygenation via the two routes along with TEX led to the best therapeutic outcome. Antitumor immune responses directed by TEX became optimized when systemic (IP) and local (IT) oxygenation was applied compared to administration of TEX alone. Results demonstrated a significant reduction in tumor size and the highest levels of IFN-γ and IL-17 and the lowest levels of IL-4 FoxP3, HIF-1α, VEGF, MMP-2, and MMP-9 in the IT+IP+TEX-treated group. Oxygenated water on the one hand could reduce tumor size, hypoxia, angiogenesis, and metastasis in the tumor microenvironment and on the other hand increases the effective immune response against the tumor systemically. This therapeutic approach is proposed as a new strategy for devising vaccines in a personalized approach.
Collapse
|
35
|
Messeha SS, Zarmouh NO, Soliman KFA. Polyphenols Modulating Effects of PD-L1/PD-1 Checkpoint and EMT-Mediated PD-L1 Overexpression in Breast Cancer. Nutrients 2021; 13:nu13051718. [PMID: 34069461 PMCID: PMC8159140 DOI: 10.3390/nu13051718] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2021] [Revised: 05/11/2021] [Accepted: 05/17/2021] [Indexed: 12/12/2022] Open
Abstract
Investigating dietary polyphenolic compounds as antitumor agents are rising due to the growing evidence of the close association between immunity and cancer. Cancer cells elude immune surveillance for enhancing their progression and metastasis utilizing various mechanisms. These mechanisms include the upregulation of programmed death-ligand 1 (PD-L1) expression and Epithelial-to-Mesenchymal Transition (EMT) cell phenotype activation. In addition to its role in stimulating normal embryonic development, EMT has been identified as a critical driver in various aspects of cancer pathology, including carcinogenesis, metastasis, and drug resistance. Furthermore, EMT conversion to another phenotype, Mesenchymal-to-Epithelial Transition (MET), is crucial in developing cancer metastasis. A central mechanism in the upregulation of PD-L1 expression in various cancer types is EMT signaling activation. In breast cancer (BC) cells, the upregulated level of PD-L1 has become a critical target in cancer therapy. Various signal transduction pathways are involved in EMT-mediated PD-L1 checkpoint overexpression. Three main groups are considered potential targets in EMT development; the effectors (E-cadherin and Vimentin), the regulators (Zeb, Twist, and Snail), and the inducers that include members of the transforming growth factor-beta (TGF-β). Meanwhile, the correlation between consuming flavonoid-rich food and the lower risk of cancers has been demonstrated. In BC, polyphenols were found to downregulate PD-L1 expression. This review highlights the effects of polyphenols on the EMT process by inhibiting mesenchymal proteins and upregulating the epithelial phenotype. This multifunctional mechanism could hold promises in the prevention and treating breast cancer.
Collapse
Affiliation(s)
- Samia S. Messeha
- Division of Pharmaceutical Sciences, College of Pharmacy & Pharmaceutical Sciences, Institute of Public Health Florida A&M University, Tallahassee, FL 32307, USA;
| | - Najla O. Zarmouh
- Faculty of Medical Technology-Misrata, Libyan National Board for Technical & Vocational Education, Misrata LY72, Libya;
| | - Karam F. A. Soliman
- Division of Pharmaceutical Sciences, College of Pharmacy & Pharmaceutical Sciences, Institute of Public Health Florida A&M University, Tallahassee, FL 32307, USA;
- Correspondence: ; Tel.: +1-850-599-3306; Fax: +1-850-599-3667
| |
Collapse
|
36
|
Druker J, Wilson JW, Child F, Shakir D, Fasanya T, Rocha S. Role of Hypoxia in the Control of the Cell Cycle. Int J Mol Sci 2021; 22:ijms22094874. [PMID: 34062959 PMCID: PMC8124716 DOI: 10.3390/ijms22094874] [Citation(s) in RCA: 24] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2021] [Revised: 05/02/2021] [Accepted: 05/03/2021] [Indexed: 12/22/2022] Open
Abstract
The cell cycle is an important cellular process whereby the cell attempts to replicate its genome in an error-free manner. As such, mechanisms must exist for the cell cycle to respond to stress signals such as those elicited by hypoxia or reduced oxygen availability. This review focuses on the role of transcriptional and post-transcriptional mechanisms initiated in hypoxia that interface with cell cycle control. In addition, we discuss how the cell cycle can alter the hypoxia response. Overall, the cellular response to hypoxia and the cell cycle are linked through a variety of mechanisms, allowing cells to respond to hypoxia in a manner that ensures survival and minimal errors throughout cell division.
Collapse
Affiliation(s)
- Jimena Druker
- Centre for Gene Regulation and Expression, School of Life Sciences, University of Dundee, Dundee DD1 5EH, UK;
| | - James W. Wilson
- Department of Molecular Physiology and Cell Signalling, Institute of Systems, Molecular and Integrative Biology, University of Liverpool, Liverpool L69 7ZB, UK; (J.W.W.); (F.C.); (D.S.); (T.F.)
| | - Fraser Child
- Department of Molecular Physiology and Cell Signalling, Institute of Systems, Molecular and Integrative Biology, University of Liverpool, Liverpool L69 7ZB, UK; (J.W.W.); (F.C.); (D.S.); (T.F.)
| | - Dilem Shakir
- Department of Molecular Physiology and Cell Signalling, Institute of Systems, Molecular and Integrative Biology, University of Liverpool, Liverpool L69 7ZB, UK; (J.W.W.); (F.C.); (D.S.); (T.F.)
| | - Temitope Fasanya
- Department of Molecular Physiology and Cell Signalling, Institute of Systems, Molecular and Integrative Biology, University of Liverpool, Liverpool L69 7ZB, UK; (J.W.W.); (F.C.); (D.S.); (T.F.)
| | - Sonia Rocha
- Department of Molecular Physiology and Cell Signalling, Institute of Systems, Molecular and Integrative Biology, University of Liverpool, Liverpool L69 7ZB, UK; (J.W.W.); (F.C.); (D.S.); (T.F.)
- Correspondence: ; Tel.: +44-(0)151-794-9084
| |
Collapse
|
37
|
Severe Acute Respiratory Syndrome Coronavirus-2-Associated Acute Kidney Injury: A Narrative Review Focused Upon Pathophysiology. Crit Care Med 2021; 49:e533-e540. [PMID: 33405411 DOI: 10.1097/ccm.0000000000004889] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023]
Abstract
OBJECTIVE Severe acute respiratory syndrome coronavirus-2 acute kidney injury is a condition that in many ways resembles classical acute kidney injury. The pathophysiology appears to be multifactorial, and accordingly, our main objective was to review possible components of this form of acute kidney injury. DATA SOURCES Literature review. DATA SYNTHESIS Our principal observation was that the various components of severe acute respiratory syndrome coronavirus-2 acute kidney injury appear to be relatively similar to the classical forms. Temporality of injury is an important factor but is not specific to severe acute respiratory syndrome coronavirus-2 acute kidney injury. Several insults hit the kidney at different moments in the course of disease, some occurring prior to hospital admission, whereas others take place at various stages during hospitalization. CONCLUSIONS AND RELEVANCE Treatment of severe acute respiratory syndrome coronavirus-2 acute kidney injury cannot be approached in a "one-size-fits-all" manner. The numerous mechanisms involved do not occur simultaneously, leading to a multiple hit model that may contribute to the prevalence and severity of acute kidney injury. A personalized approach to each patient with acute kidney injury based on the timing and severity of disease is necessary in order to provide appropriate treatment. Although data from the literature regarding the previous coronavirus infections can give some insights, more studies are needed to explore the different mechanisms of acute kidney injury occurring as a result of severe acute respiratory syndrome coronavirus-2.
Collapse
|
38
|
Zhang L, Zhao Y, Tu Q, Xue X, Zhu X, Zhao KN. The Roles of Programmed Cell Death Ligand-1/ Programmed Cell Death-1 (PD-L1/PD-1) in HPV-induced Cervical Cancer and Potential for their Use in Blockade Therapy. Curr Med Chem 2021; 28:893-909. [PMID: 32003657 DOI: 10.2174/0929867327666200128105459] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2019] [Revised: 12/13/2019] [Accepted: 12/14/2019] [Indexed: 11/22/2022]
Abstract
BACKGROUND Cervical cancer induced by infection with human papillomavirus (HPV) remains a leading cause of mortality for women worldwide although preventive vaccines and early diagnosis have reduced morbidity and mortality. Advanced cervical cancer can only be treated with either chemotherapy or radiotherapy but the outcomes are poor. The median survival for advanced cervical cancer patients is only 16.8 months. METHODS We undertook a structural search of peer-reviewed published studies based on 1). Characteristics of programmed cell death ligand-1/programmed cell death-1(PD-L1/PD-1) expression in cervical cancer and upstream regulatory signals of PD-L1/PD-1 expression, 2). The role of the PD-L1/PD-1 axis in cervical carcinogenesis induced by HPV infection and 3). Whether the PD-L1/PD-1 axis has emerged as a potential target for cervical cancer therapies. RESULTS One hundred and twenty-six published papers were included in the review, demonstrating that expression of PD-L1/PD-1 is associated with HPV-caused cancer, especially with HPV 16 and 18 which account for approximately 70% of cervical cancer cases. HPV E5/E6/E7 oncogenes activate multiple signalling pathways including PI3K/AKT, MAPK, hypoxia-inducible factor 1α, STAT3/NF-kB and microRNA, which regulate PD-L1/PD-1 axis to promote HPV-induced cervical carcinogenesis. The PD-L1/PD-1 axis plays a crucial role in the immune escape of cervical cancer through inhibition of host immune response. Creating an "immune-privileged" site for initial viral infection and subsequent adaptive immune resistance, which provides a rationale for the therapeutic blockade of this axis in HPV-positive cancers. Currently, Phase I/II clinical trials evaluating the effects of PDL1/ PD-1 targeted therapies are in progress for cervical carcinoma, which provide an important opportunity for the application of anti-PD-L1/anti-PD-1 antibodies in cervical cancer treatment. CONCLUSION Recent research developments have led to an entirely new class of drugs using antibodies against the PD-L1/PD-1 thus promoting the body's immune system to fight cancer. The expression and roles of the PD-L1/ PD-1 axis in the progression of cervical cancer provide great potential for using PD-L1/PD-1 antibodies as a targeted cancer therapy.
Collapse
Affiliation(s)
- Lifang Zhang
- School of Basic Medical Science, Wenzhou Medical University, Wenzhou, 325035 Zhejiang, China
| | - Yu Zhao
- Department of Obstetrics and Gynaecology, The Second Affiliated Hospital of Wenzhou Medical University, Wenzhou 325035, Zhejiang, China
| | - Quanmei Tu
- Department of Obstetrics and Gynaecology, The Second Affiliated Hospital of Wenzhou Medical University, Wenzhou 325035, Zhejiang, China
| | - Xiangyang Xue
- School of Basic Medical Science, Wenzhou Medical University, Wenzhou, 325035 Zhejiang, China
| | - Xueqiong Zhu
- Department of Obstetrics and Gynaecology, The Second Affiliated Hospital of Wenzhou Medical University, Wenzhou 325035, Zhejiang, China
| | - Kong-Nan Zhao
- School of Basic Medical Science, Wenzhou Medical University, Wenzhou, 325035 Zhejiang, China
| |
Collapse
|
39
|
Resistance of Hypoxic Cells to Ionizing Radiation Is Mediated in Part via Hypoxia-Induced Quiescence. Cells 2021; 10:cells10030610. [PMID: 33801903 PMCID: PMC7998378 DOI: 10.3390/cells10030610] [Citation(s) in RCA: 17] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2021] [Revised: 03/06/2021] [Accepted: 03/08/2021] [Indexed: 12/19/2022] Open
Abstract
Double strand breaks (DSBs) are highly toxic to a cell, a property that is exploited in radiation therapy. A critical component for the damage induction is cellular oxygen, making hypoxic tumor areas refractory to the efficacy of radiation treatment. During a fractionated radiation regimen, these hypoxic areas can be re-oxygenated. Nonetheless, hypoxia still constitutes a negative prognostic factor for the patient’s outcome. We hypothesized that this might be attributed to specific hypoxia-induced cellular traits that are maintained upon reoxygenation. Here, we show that reoxygenation of hypoxic non-transformed RPE-1 cells fully restored induction of DSBs but the cells remain radioresistant as a consequence of hypoxia-induced quiescence. With the use of the cell cycle indicators (FUCCI), cell cycle-specific radiation sensitivity, the cell cycle phase duration with live cell imaging, and single cell tracing were assessed. We observed that RPE-1 cells experience a longer G1 phase under hypoxia and retain a large fraction of cells that are non-cycling. Expression of HPV oncoprotein E7 prevents hypoxia-induced quiescence and abolishes the radioprotective effect. In line with this, HPV-negative cancer cell lines retain radioresistance, while HPV-positive cancer cell lines are radiosensitized upon reoxygenation. Quiescence induction in hypoxia and its HPV-driven prevention was observed in 3D multicellular spheroids. Collectively, we identify a new hypoxia-dependent radioprotective phenotype due to hypoxia-induced quiescence that accounts for a global decrease in radiosensitivity that can be retained upon reoxygenation and is absent in cells expressing oncoprotein E7.
Collapse
|
40
|
Markolin P, Davidson N, Hirt CK, Chabbert CD, Zamboni N, Schwank G, Krek W, Rätsch G. Identification of HIF-dependent alternative splicing in gastrointestinal cancers and characterization of a long, coding isoform of SLC35A3. Genomics 2021; 113:515-529. [PMID: 33418078 DOI: 10.1016/j.ygeno.2020.12.039] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2020] [Revised: 12/07/2020] [Accepted: 12/28/2020] [Indexed: 12/30/2022]
Abstract
Intra-tumor hypoxia is a common feature in many solid cancers. Although transcriptional targets of hypoxia-inducible factors (HIFs) have been well characterized, alternative splicing or processing of pre-mRNA transcripts which occurs during hypoxia and subsequent HIF stabilization is much less understood. Here, we identify many HIF-dependent alternative splicing events after whole transcriptome sequencing in pancreatic cancer cells exposed to hypoxia with and without downregulation of the aryl hydrocarbon receptor nuclear translocator (ARNT), a protein required for HIFs to form a transcriptionally active dimer. We correlate the discovered hypoxia-driven events with available sequencing data from pan-cancer TCGA patient cohorts to select a narrow set of putative biologically relevant splice events for experimental validation. We validate a small set of candidate HIF-dependent alternative splicing events in multiple human gastrointestinal cancer cell lines as well as patient-derived human pancreatic cancer organoids. Lastly, we report the discovery of a HIF-dependent mechanism to produce a hypoxia-dependent, long and coding isoform of the UDP-N-acetylglucosamine transporter SLC35A3.
Collapse
Affiliation(s)
- Philipp Markolin
- Institute of Molecular Health Sciences, ETH Zurich, 8093 Zurich, Switzerland; Biomedical Informatics Group, ETH Zurich, 8092 Zürich, Switzerland
| | - Natalie Davidson
- Biomedical Informatics Group, ETH Zurich, 8092 Zürich, Switzerland
| | - Christian K Hirt
- Institute of Molecular Health Sciences, ETH Zurich, 8093 Zurich, Switzerland
| | | | - Nicola Zamboni
- Institute of Molecular Systems Biology, ETH Zurich, 8093 Zürich, Switzerland
| | - Gerald Schwank
- Institute of Molecular Health Sciences, ETH Zurich, 8093 Zurich, Switzerland; Biomedical Informatics Group, ETH Zurich, 8092 Zürich, Switzerland
| | - Wilhelm Krek
- Institute of Molecular Health Sciences, ETH Zurich, 8093 Zurich, Switzerland
| | - Gunnar Rätsch
- Biomedical Informatics Group, ETH Zurich, 8092 Zürich, Switzerland.
| |
Collapse
|
41
|
Younis I. Dehisced abdominal wall reconstruction. J Wound Care 2021; 29:S29-S30. [PMID: 32427032 DOI: 10.12968/jowc.2020.29.sup5b.s29] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/11/2022]
Affiliation(s)
- Ibby Younis
- Consultant Plastic and Reconstructive Surgeon, Royal Free London NHS Foundation Trust, London, UK
| |
Collapse
|
42
|
Jha PK, Vijay A, Prabhakar A, Chatterjee T, Nair V, Bajaj N, Kumar B, Sharma M, Ashraf MZ. Transcriptome Profiling Reveals the Endogenous Sponging Role of LINC00659 and UST-AS1 in High-Altitude Induced Thrombosis. Thromb Haemost 2021; 121:1497-1511. [PMID: 33580494 DOI: 10.1055/a-1390-1713] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
BACKGROUND The pathophysiology of deep vein thrombosis (DVT) is considered as multifactorial, where thrombus formation is an interplay of genetic and acquired risk factors. Little is known about the expression profile and roles of long noncoding RNAs (lncRNAs) in human subjects developing DVT at high altitude. METHODS Using RNAseQ, we compared peripheral blood mRNA and lncRNA expression profile in human high-altitude DVT (HA-DVT) patients with high-altitude control subjects. We used DESeq to identify differentially expressed (DE) genes. We annotated the lncRNAs using NONCODE 3.0 database. In silico putative lncRNA-miRNA association study unravels the endogenous miRNA sponge associated with our candidate lncRNAs. These findings were validated by small-interfering RNA (siRNA) knockdown assay of the candidate lncRNAs conducted in primary endothelial cells. RESULTS We identified 1,524 DE mRNAs and 973 DE lncRNAs. Co-expressed protein-coding gene analysis resulted in a list of 722 co-expressed protein-coding genes with a Pearson correlation coefficients >0.7. The functional annotation of co-expressed genes and putative proteins revealed their involvement in the hypoxia, immune response, and coagulation cascade. Through its miRNA response elements to compete for miR-143 and miR-15, lncRNA-LINC00659 and UXT-AS1 regulate the expression of prothrombotic genes. Furthermore, in vitro RNA interference (siRNA) simultaneously suppressed lncRNAs and target gene mRNA level. CONCLUSION This transcriptome profile describes novel potential mechanisms of interaction between lncRNAs, the coding genes, miRNAs, and regulatory transcription factors that define the thrombotic signature and may be used in establishing lncRNAs as a biomarker in HA-DVT.
Collapse
Affiliation(s)
- Prabhash Kumar Jha
- Defence Institute of Physiology and Allied Sciences, Defence Research and Development Organisation, Delhi, India
| | - Aatira Vijay
- Defence Institute of Physiology and Allied Sciences, Defence Research and Development Organisation, Delhi, India
| | - Amit Prabhakar
- Defence Institute of Physiology and Allied Sciences, Defence Research and Development Organisation, Delhi, India
| | | | - Velu Nair
- Armed Forces Medical College, Pune, Maharashtra, India
| | - Nitin Bajaj
- Command Hospital (Western Command), Chandimandir, Chandigarh, India
| | - Bhuvnesh Kumar
- Defence Institute of Physiology and Allied Sciences, Defence Research and Development Organisation, Delhi, India
| | - Manish Sharma
- Defence Institute of Physiology and Allied Sciences, Defence Research and Development Organisation, Delhi, India
| | - Mohammad Zahid Ashraf
- Defence Institute of Physiology and Allied Sciences, Defence Research and Development Organisation, Delhi, India
| |
Collapse
|
43
|
Oxidative Stress-Part of the Solution or Part of the Problem in the Hypoxic Environment of a Brain Tumor. Antioxidants (Basel) 2020; 9:antiox9080747. [PMID: 32823815 PMCID: PMC7464568 DOI: 10.3390/antiox9080747] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2020] [Revised: 08/08/2020] [Accepted: 08/12/2020] [Indexed: 12/13/2022] Open
Abstract
Rapid growth of brain tumors such as glioblastoma often results in oxygen deprivation and the emergence of hypoxic zones. In consequence, the enrichment of reactive oxygen species occurs, harming nonmalignant cells and leading them toward apoptotic cell death. However, cancer cells survive such exposure and thrive in a hypoxic environment. As the mechanisms responsible for such starkly different outcomes are not sufficiently explained, we aimed to explore what transcriptome rearrangements are used by glioblastoma cells in hypoxic areas. Using metadata analysis of transcriptome in different subregions of the glioblastoma retrieved from the Ivy Glioblastoma Atlas Project, we created the reactive oxygen species-dependent map of the transcriptome. This map was then used for the analysis of differential gene expression in the histologically determined cellular tumors and hypoxic zones. The gene ontology analysis cross-referenced with the clinical data from The Cancer Genome Atlas revealed that the metabolic shift is one of the major prosurvival strategies applied by cancer cells to overcome hypoxia-related cytotoxicity.
Collapse
|
44
|
Abbasi A, Pakravan N, Hassan ZM. Hyaluronic acid optimises therapeutic effects of hydrogen peroxide-induced oxidative stress on breast cancer. J Cell Physiol 2020; 236:1494-1514. [PMID: 32740942 DOI: 10.1002/jcp.29957] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2020] [Accepted: 07/08/2020] [Indexed: 12/20/2022]
Abstract
Distinguishing the multiple effects of reactive oxygen species (ROS) on cancer cells is important to understand their role in tumour biology. On one side, ROS can be oncogenic by promoting hypoxic conditions, genomic instability and tumorigenesis. Conversely, elevated levels of ROS-induced oxidative stress can induce cancer cell death. This is evidenced by the conflicting results of research using antioxidant therapy, which in some cases promoted tumour growth and metastasis. However, some antioxidative or ROS-mediated oxidative therapies have also yielded beneficial effects. To better define the effects of oxidative stress, in vitro experiments were conducted on 4T1 and splenic mononuclear cells (MNCs) under hypoxic and normoxic conditions. Furthermore, hydrogen peroxide (H2 O2 ; 10-1,000 μM) was used as an ROS source alone or in combination with hyaluronic acid (HA), which is frequently used as drug delivery vehicle. Our result indicated that the treatment of cancer cells with H2 O2 + HA was significantly more effective than H2 O2 alone. In addition, treatment with H2 O2 + HA led to increased apoptosis, decreased proliferation, and multiphase cell cycle arrest in 4T1 cells in a dose-dependent manner under normoxic or hypoxic conditions. As a result, migratory tendency and the messenger RNA levels of vascular endothelial growth factor, matrix metalloproteinase-2 (MMP-2), and MMP-9 were significantly decreased in 4T1 cells. Of note, HA treatment combined with 100-1,000 μM H2 O2 caused more damage to MNCs as compared to treatment with lower concentrations (10-50 μM). Based on these results, we propose to administer high-dose H2 O2 + HA (100-1000 μM) for intratumoural injection and low doses for systemic administration. Intratumoural route could have toxic and inhibitory effects not only on the tumour but also on residential myeloid cells defending it, whereas systemic treatment could stimulate peripheral immune responses against the tumour. More in vivo research is required to confirm this hypothesis.
Collapse
Affiliation(s)
- Ardeshir Abbasi
- Department of Immunology, Faculty of Medical Sciences, Tarbiat Modares University, Tehran, Iran
| | - Nafiseh Pakravan
- Department of Immunology, Medical School, Alborz University of Medical Sciences, Karaj, Iran
| | - Zuhair Mohammad Hassan
- Department of Immunology, Faculty of Medical Sciences, Tarbiat Modares University, Tehran, Iran
| |
Collapse
|
45
|
Nguyen VT, Nardini M, Ruggiu A, Cancedda R, Descalzi F, Mastrogiacomo M. Platelet Lysate Induces in Human Osteoblasts Resumption of Cell Proliferation and Activation of Pathways Relevant for Revascularization and Regeneration of Damaged Bone. Int J Mol Sci 2020; 21:ijms21145123. [PMID: 32698534 PMCID: PMC7403959 DOI: 10.3390/ijms21145123] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2020] [Revised: 07/13/2020] [Accepted: 07/16/2020] [Indexed: 12/17/2022] Open
Abstract
To understand the regenerative effect of platelet-released molecules in bone repair one should investigate the cascade of events involving the resident osteoblast population during the reconstructive process. Here the in vitro response of human osteoblasts to a platelet lysate (PL) stimulus is reported. Quiescent or very slow dividing osteoblasts showed a burst of proliferation after PL stimulation and returned to a none or very slow dividing condition when the PL was removed. PL stimulated osteoblasts maintained a differentiation capability in vitro and in vivo when tested in absence of PL. Since angiogenesis plays a crucial role in the bone healing process, we investigated in PL stimulated osteoblasts the activation of hypoxia-inducible factor 1-alpha (HIF-1α) and signal transducer and activator of transcription 3 (STAT3) pathways, involved in both angiogenesis and bone regeneration. We observed phosphorylation of STAT3 and a strong induction, nuclear translocation and DNA binding of HIF-1α. In agreement with the induction of HIF-1α an enhanced secretion of vascular endothelial growth factor (VEGF) occurred. The double effect of the PL on quiescent osteoblasts, i.e., resumption of proliferation and activation of pathways promoting both angiogenesis and bone formation, provides a rationale to the application of PL as therapeutic agent in post-traumatic bone repair.
Collapse
Affiliation(s)
- Van Thi Nguyen
- Department of Experimental Medicine (DIMES), University of Genova, 16132 Genova, Italy; (V.T.N.); (A.R.); (F.D.)
| | - Marta Nardini
- Department of Internal Medicine (DIMI), University of Genova, 16132 Genova, Italy;
- Biotherapy Unit, Ospedale Policlinico San Martino, 16132 Genova, Italy
| | - Alessandra Ruggiu
- Department of Experimental Medicine (DIMES), University of Genova, 16132 Genova, Italy; (V.T.N.); (A.R.); (F.D.)
| | | | - Fiorella Descalzi
- Department of Experimental Medicine (DIMES), University of Genova, 16132 Genova, Italy; (V.T.N.); (A.R.); (F.D.)
| | - Maddalena Mastrogiacomo
- Department of Internal Medicine (DIMI), University of Genova, 16132 Genova, Italy;
- Biotherapy Unit, Ospedale Policlinico San Martino, 16132 Genova, Italy
- Center for Biomedical Research (CEBR), University of Genova, 16132 Genova, Italy
- Correspondence: ; Tel.: +39-010-555-8203
| |
Collapse
|
46
|
Kareliotis G, Tremi I, Kaitatzi M, Drakaki E, Serafetinides AA, Makropoulou M, Georgakilas AG. Combined radiation strategies for novel and enhanced cancer treatment. Int J Radiat Biol 2020; 96:1087-1103. [PMID: 32602416 DOI: 10.1080/09553002.2020.1787544] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022]
Abstract
Numerous studies focus on cancer therapy worldwide, and although many advances have been recorded, the complexity of the disease dictates thinking out of the box to confront it. This study reviews some of the currently available ionizing (IR) and non-ionizing radiation (NIR)-based treatment methods and explores their possible combinations that lead to synergistic, multimodal approaches with promising therapeutic outcomes. Traditional techniques, like radiotherapy (RT) show decent results, although they cannot spare 100% the healthy tissues neighboring with the cancer ones. Targeted therapies, such as proton and photodynamic therapy (PT and PDT, respectively) present adequate outcomes, even though each one has its own drawbacks. To overcome these limitations, the combination of therapeutic modalities has been proposed and has already been showing promising results. At the same time, the recent advances in nanotechnology in the form of nanoparticles enhance cancer therapy, making multimodal treatments worthy of exploring and studying. The combination of RT and PDT has reached the level of clinical trials and is showing promising results. Moreover, in vitro and in vivo studies of nanoparticles with PDT have also provided beneficial results concerning enhanced radiation treatments. In any case, novel and multimodal approaches have to be adopted to achieve personalized, enhanced and effective cancer treatment.
Collapse
Affiliation(s)
- Georgios Kareliotis
- Department of Physics, School of Applied Mathematical and Physical Sciences, National Technical University of Athens (NTUA), Athens, Greece
| | - Ioanna Tremi
- Department of Physics, School of Applied Mathematical and Physical Sciences, National Technical University of Athens (NTUA), Athens, Greece
| | - Myrsini Kaitatzi
- Department of Physics, School of Applied Mathematical and Physical Sciences, National Technical University of Athens (NTUA), Athens, Greece
| | - Eleni Drakaki
- Department of Physics, School of Applied Mathematical and Physical Sciences, National Technical University of Athens (NTUA), Athens, Greece
| | - Alexandros A Serafetinides
- Department of Physics, School of Applied Mathematical and Physical Sciences, National Technical University of Athens (NTUA), Athens, Greece
| | - Mersini Makropoulou
- Department of Physics, School of Applied Mathematical and Physical Sciences, National Technical University of Athens (NTUA), Athens, Greece
| | - Alexandros G Georgakilas
- Department of Physics, School of Applied Mathematical and Physical Sciences, National Technical University of Athens (NTUA), Athens, Greece
| |
Collapse
|
47
|
Luo A, Gong Y, Kim H, Chen Y. Proteome dynamics analysis identifies functional roles of SDE2 and hypoxia in DNA damage response in prostate cancer cells. NAR Cancer 2020; 2:zcaa010. [PMID: 32743553 PMCID: PMC7380487 DOI: 10.1093/narcan/zcaa010] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2020] [Revised: 05/19/2020] [Accepted: 06/08/2020] [Indexed: 12/20/2022] Open
Abstract
Mechanistic understanding of hypoxia-responsive signaling pathways provides important insights into oxygen- and metabolism-dependent cellular phenotypes in diseases. Using SILAC-based quantitative proteomics, we provided a quantitative map identifying over 6300 protein groups in response to hypoxia in prostate cancer cells and identified both canonical and novel cellular networks dynamically regulated under hypoxia. Particularly, we identified SDE2, a DNA stress response modulator, that was significantly downregulated by hypoxia, independent of HIF (hypoxia-inducible factor) transcriptional activity. Mechanistically, hypoxia treatment promoted SDE2 polyubiquitination and degradation. Such regulation is independent of previously identified Arg/N-end rule proteolysis or the ubiquitin E3 ligase, CDT2. Depletion of SDE2 increased cellular sensitivity to DNA damage and inhibited cell proliferation. Interestingly, either SDE2 depletion or hypoxia treatment potentiated DNA damage-induced PCNA (proliferating cell nuclear antigen) monoubiquitination, a key step for translesion DNA synthesis. Furthermore, knockdown of SDE2 desensitized, while overexpression of SDE2 protected the hypoxia-mediated regulation of PCNA monoubiquitination upon DNA damage. Taken together, our quantitative proteomics and biochemical study revealed diverse hypoxia-responsive pathways that strongly associated with prostate cancer tumorigenesis and identified the functional roles of SDE2 and hypoxia in regulating DNA damage-induced PCNA monoubiquitination, suggesting a possible link between hypoxic microenvironment and the activation of error-prone DNA repair pathway in tumor cells.
Collapse
Affiliation(s)
- Ang Luo
- Department of Biochemistry, Molecular Biology and Biophysics, University of Minnesota at Twin Cities, Minneapolis, MN 55455, USA
| | - Yao Gong
- Department of Biochemistry, Molecular Biology and Biophysics, University of Minnesota at Twin Cities, Minneapolis, MN 55455, USA
| | - Hyungjin Kim
- Department of Pharmacological Sciences, Stony Brook University, Stony Brook, NY 11794, USA
| | - Yue Chen
- Department of Biochemistry, Molecular Biology and Biophysics, University of Minnesota at Twin Cities, Minneapolis, MN 55455, USA
| |
Collapse
|
48
|
Wilson JW, Shakir D, Batie M, Frost M, Rocha S. Oxygen-sensing mechanisms in cells. FEBS J 2020; 287:3888-3906. [PMID: 32446269 DOI: 10.1111/febs.15374] [Citation(s) in RCA: 42] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2020] [Revised: 04/24/2020] [Accepted: 05/11/2020] [Indexed: 12/15/2022]
Abstract
The importance of oxygen for the survival of multicellular and aerobic organisms is well established and documented. Over the years, increased knowledge of its use for bioenergetics has placed oxygen at the centre of research on mitochondria and ATP-generating processes. Understanding the molecular mechanisms governing cellular oxygen sensing and response has allowed for the discovery of novel pathways oxygen is involved in, culminating with the award of the Nobel Prize for Medicine and Physiology in 2019 to the pioneers of this field, Greg Semenza, Peter Ratcliffe and William Kaelin. However, it is now beginning to be appreciated that oxygen can be a signalling molecule involved in a vast array of molecular processes, most of which impinge on gene expression control. This review will focus on the knowns and unknowns of oxygen as a signalling molecule, highlighting the role of 2-oxoglutarate-dependent dioxygenases as central players in the cellular response to deviations in oxygen tension.
Collapse
Affiliation(s)
- James W Wilson
- Department of Biochemistry, Institute of Integrative Biology, University of Liverpool, UK
| | - Dilem Shakir
- Department of Biochemistry, Institute of Integrative Biology, University of Liverpool, UK
| | - Michael Batie
- Department of Biochemistry, Institute of Integrative Biology, University of Liverpool, UK
| | - Mark Frost
- Department of Biochemistry, Institute of Integrative Biology, University of Liverpool, UK
| | - Sonia Rocha
- Department of Biochemistry, Institute of Integrative Biology, University of Liverpool, UK
| |
Collapse
|
49
|
Sofer T, Li R, Joehanes R, Lin H, Gower AC, Wang H, Kurniansyah N, Cade BE, Lee J, Williams S, Mehra R, Patel SR, Quan SF, Liu Y, Rotter JI, Rich SS, Spira A, Levy D, Gharib SA, Redline S, Gottlieb DJ. Low oxygen saturation during sleep reduces CD1D and RAB20 expressions that are reversed by CPAP therapy. EBioMedicine 2020; 56:102803. [PMID: 32512511 PMCID: PMC7276515 DOI: 10.1016/j.ebiom.2020.102803] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2020] [Revised: 04/13/2020] [Accepted: 05/05/2020] [Indexed: 02/08/2023] Open
Abstract
BACKGROUND Sleep Disordered Breathing (SDB) is associated with a wide range of pathophysiological changes due, in part, to hypoxemia during sleep. We sought to identify gene transcription associations with measures of SDB and hypoxemia during sleep, and study their response to treatment. METHODS In two discovery cohorts, Framingham Offspring Study (FOS; N = 571) and the Multi-Ethnic Study of Atherosclerosis (MESA; N = 580), we studied gene expression in peripheral blood mononuclear cells in association with three measures of SDB: Apnea Hypopnea Index (AHI); average oxyhemoglobin saturation (avgO2) during sleep; and minimum oxyhemoglobin saturation (minO2) during sleep. Associated genes were used for analysis of gene expression in the blood of 15 participants with moderate or severe obstructive sleep apnea (OSA) from the Heart Biomarkers In Apnea Treatment (HeartBEAT) trial. These genes were studied pre- and post-treatment (three months) with continuous positive airway pressure (CPAP). We also performed Gene Set Enrichment Analysis (GSEA) on all traits and cohort analyses. FINDINGS Twenty-two genes were associated with SDB traits in both MESA and FOS. Of these, lower expression of CD1D and RAB20 was associated with lower avgO2 in MESA and FOS. CPAP treatment increased the expression of these genes in HeartBEAT participants. Immunity and inflammation pathways were up-regulated in subjects with lower avgO2; i.e., in those with a more severe SDB phenotype (MESA), whereas immuno-inflammatory processes were down-regulated following CPAP treatment (HeartBEAT). INTERPRETATION Low oxygen saturation during sleep is associated with alterations in gene expression and transcriptional programs that are partially reversed by CPAP treatment.
Collapse
Affiliation(s)
- Tamar Sofer
- Division of Sleep Medicine, Harvard Medical School, Boston, MA 02115, USA; Division of Sleep and Circadian Disorders, Department of Medicine, Brigham and Women's Hospital, Boston, MA, USA; Department of Biostatistics, Harvard T. H. Chan School of Public Health, Boston, MA, USA.
| | - Ruitong Li
- Division of Sleep and Circadian Disorders, Department of Medicine, Brigham and Women's Hospital, Boston, MA, USA
| | - Roby Joehanes
- The Population Sciences Branch of the National Heart, Lung and Blood Institute, Bethesda, MD, USA and the Framingham Heart Study, Framingham, MA, USA; Hebrew SeniorLife, Harvard Medical School, Boston, MA, USA
| | - Honghuang Lin
- The Population Sciences Branch of the National Heart, Lung and Blood Institute, Bethesda, MD, USA and the Framingham Heart Study, Framingham, MA, USA; Department of Medicine, Boston University School of Medicine, Boston, MA, USA
| | - Adam C Gower
- Clinical and Translational Science Institute, Boston University School of Medicine, Boston, USA
| | - Heming Wang
- Division of Sleep Medicine, Harvard Medical School, Boston, MA 02115, USA; Division of Sleep and Circadian Disorders, Department of Medicine, Brigham and Women's Hospital, Boston, MA, USA
| | - Nuzulul Kurniansyah
- Division of Sleep and Circadian Disorders, Department of Medicine, Brigham and Women's Hospital, Boston, MA, USA
| | - Brian E Cade
- Division of Sleep Medicine, Harvard Medical School, Boston, MA 02115, USA; Division of Sleep and Circadian Disorders, Department of Medicine, Brigham and Women's Hospital, Boston, MA, USA
| | - Jiwon Lee
- Division of Sleep and Circadian Disorders, Department of Medicine, Brigham and Women's Hospital, Boston, MA, USA
| | - Stephanie Williams
- Division of Sleep and Circadian Disorders, Department of Medicine, Brigham and Women's Hospital, Boston, MA, USA
| | - Reena Mehra
- Neurologic Institute, Cleveland Clinic, Cleveland, OH, USA
| | - Sanjay R Patel
- Division of Pulmonary, Allergy and Critical Care Medicine, University of Pittsburgh, Pittsburgh, PA, USA
| | - Stuart F Quan
- Division of Sleep Medicine, Harvard Medical School, Boston, MA 02115, USA; Division of Sleep and Circadian Disorders, Department of Medicine, Brigham and Women's Hospital, Boston, MA, USA
| | - Yongmei Liu
- Department of Medicine, Duke University School of Medicine, Durham, NC, USA
| | - Jerome I Rotter
- The Institute for Translational Genomics and Population Sciences, Departments of Pediatrics and Medicine, Los Angeles Biomedical Research Institute at Harbor-UCLA Medical Center, Torrance, CA, USA
| | - Stephen S Rich
- Center for Public Health Genomics, Department of Public Health Sciences, University of Virginia, Charlottesville, VA, USA
| | - Avrum Spira
- Boston University School of Medicine and Boston Medical Center, Boston, MA, USA
| | - Daniel Levy
- The Population Sciences Branch of the National Heart, Lung and Blood Institute, Bethesda, MD, USA and the Framingham Heart Study, Framingham, MA, USA
| | - Sina A Gharib
- Computational Medicine Core, Center for Lung Biology, University of Washington Medicine Sleep Center, Division of Pulmonary, Critical Care, and Sleep Medicine, University of Washington, Seattle, WA, USA
| | - Susan Redline
- Division of Sleep Medicine, Harvard Medical School, Boston, MA 02115, USA; Division of Sleep and Circadian Disorders, Department of Medicine, Brigham and Women's Hospital, Boston, MA, USA; Computational Medicine Core, Center for Lung Biology, University of Washington Medicine Sleep Center, Division of Pulmonary, Critical Care, and Sleep Medicine, University of Washington, Seattle, WA, USA
| | - Daniel J Gottlieb
- Division of Sleep Medicine, Harvard Medical School, Boston, MA 02115, USA; Division of Sleep and Circadian Disorders, Department of Medicine, Brigham and Women's Hospital, Boston, MA, USA; Division of Pulmonary, Critical Care, and Sleep Medicine, Beth Israel Deaconess Medical Center, Boston, MA, USA; VA Boston Healthcare System, Boston, MA, USA
| |
Collapse
|
50
|
Nair HKR. Non-healing venous leg ulcer. J Wound Care 2020; 29:S26-S27. [DOI: 10.12968/jowc.2020.29.sup5b.s26] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/11/2022]
|