1
|
Liu MX, Liu YC, Cai YT, Gu YY, Zhu YQ, Zhang N, Zhu WZ, Liu YH, Yu L, Zhang QT, Zhang XL. Self-Produced O 2 CNs-Based Nanocarriers of DNA Hydrophobization Strategy Triggers Photodynamic and Mitochondrial-Derived Ferroptosis for Hepatocellular Carcinoma Combined Treatment. Adv Healthc Mater 2024; 13:e2402110. [PMID: 39205543 DOI: 10.1002/adhm.202402110] [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: 06/07/2024] [Revised: 08/17/2024] [Indexed: 09/04/2024]
Abstract
Hypoxia can aggravate tumor occurrence, development, invasion, and metastasis, and greatly inhibit the photodynamic therapy (PDT) effect. Herein, carbon nitride (CNs)-based DNA and photosensitizer co-delivery systems (BPSCNs) with oxygen-producing functions are developed to address this problem. Selenide glucose (Seglu) is used as the dopant to prepare red/NIR-active CNs (SegluCNs). The tumor-targeting unit Bio-PEG2000 is utilized to construct BPSCNs nanoparticles through esterification reactions. Furthermore, DNA hydrophobization is realized via mixing P53 gene with a positively charged mitochondrial-targeted near-infrared (NIR) emitting photosensitizer (MTTPY), which is encapsulated in non-cationic BPSCNs for synergistic delivery. Ester bonds in BPSCNs@MTTPY-P53 complexes can be disrupted by lipase in the liver to facilitate P53 release, upregulated P53 expression, and promoted HIF-1α degradation in mitochondria. In addition, the oxygen produced by the complexes improved the hypoxic microenvironment of hepatocellular carcinoma (HCC), synergistically downregulated HIF-1α expression in mitochondria, promoted mitochondrial-derived ferroptosis and enhanced the PDT effect of the MTTPY unit. Both in vivo and in vitro experiments indicated that the transfected P53-DNA, produced O2 and ROS by these complexes synergistically led to mitochondrial-derived ferroptosis in hepatoma cells through the HIF-1α/SLC7A11 pathway, and completely avoiding PDT resistance caused by hypoxia, exerting a significant therapeutic role in HCC treatment.
Collapse
Affiliation(s)
- Ming-Xuan Liu
- School of Pharmacy, Nantong University, Nantong, 226001, P. R. China
| | - Yan-Chao Liu
- School of Chemistry and Chemical Engineering, Yangzhou University, Yangzhou, Jiangsu, 225001, P. R. China
- Institute of Translational Medicine, Medical College, Yangzhou University, Yangzhou, Jiangsu, 225001, P. R. China
| | - Yu-Ting Cai
- School of Pharmacy, Nantong University, Nantong, 226001, P. R. China
| | - Ying-Ying Gu
- School of Pharmacy, Nantong University, Nantong, 226001, P. R. China
| | - Ya-Qi Zhu
- School of Pharmacy, Nantong University, Nantong, 226001, P. R. China
| | - Nan Zhang
- School of Pharmacy, Nantong University, Nantong, 226001, P. R. China
| | - Wei-Zhong Zhu
- School of Pharmacy, Nantong University, Nantong, 226001, P. R. China
| | - Yong-Hong Liu
- School of Chemistry and Chemical Engineering, Yangzhou University, Yangzhou, Jiangsu, 225001, P. R. China
- Institute of Translational Medicine, Medical College, Yangzhou University, Yangzhou, Jiangsu, 225001, P. R. China
| | - Lei Yu
- School of Chemistry and Chemical Engineering, Yangzhou University, Yangzhou, Jiangsu, 225001, P. R. China
- Institute of Translational Medicine, Medical College, Yangzhou University, Yangzhou, Jiangsu, 225001, P. R. China
| | - Qi-Tao Zhang
- International Collaborative Laboratory of 2D Materials for Optoelectronics Science and Technology of Ministry of Education, Institute of Microscale Optoelectronics, Shenzhen University, Shenzhen, 518060, P. R. China
| | - Xiao-Ling Zhang
- School of Pharmacy, Nantong University, Nantong, 226001, P. R. China
| |
Collapse
|
2
|
Quail DF, Park M, Welm AL, Ekiz HA. Breast Cancer Immunity: It is TIME for the Next Chapter. Cold Spring Harb Perspect Med 2024; 14:a041324. [PMID: 37188526 PMCID: PMC10835621 DOI: 10.1101/cshperspect.a041324] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/17/2023]
Abstract
Our ability to interrogate the tumor immune microenvironment (TIME) at an ever-increasing granularity has uncovered critical determinants of disease progression. Not only do we now have a better understanding of the immune response in breast cancer, but it is becoming possible to leverage key mechanisms to effectively combat this disease. Almost every component of the immune system plays a role in enabling or inhibiting breast tumor growth. Building on early seminal work showing the involvement of T cells and macrophages in controlling breast cancer progression and metastasis, single-cell genomics and spatial proteomics approaches have recently expanded our view of the TIME. In this article, we provide a detailed description of the immune response against breast cancer and examine its heterogeneity in disease subtypes. We discuss preclinical models that enable dissecting the mechanisms responsible for tumor clearance or immune evasion and draw parallels and distinctions between human disease and murine counterparts. Last, as the cancer immunology field is moving toward the analysis of the TIME at the cellular and spatial levels, we highlight key studies that revealed previously unappreciated complexity in breast cancer using these technologies. Taken together, this article summarizes what is known in breast cancer immunology through the lens of translational research and identifies future directions to improve clinical outcomes.
Collapse
Affiliation(s)
- Daniela F Quail
- Rosalind and Morris Goodman Cancer Institute, McGill University, Montreal, Quebec H3A 1A3, Canada
- Department of Physiology, McGill University, Montreal, Quebec H3G 1Y6, Canada
| | - Morag Park
- Rosalind and Morris Goodman Cancer Institute, McGill University, Montreal, Quebec H3A 1A3, Canada
- Departments of Biochemistry, Oncology, McGill University, Montreal, Quebec H3G 1Y6, Canada
| | - Alana L Welm
- Department of Oncological Sciences, Huntsman Cancer Institute, University of Utah, Salt Lake City, Utah 84112, USA
| | - H Atakan Ekiz
- Department of Molecular Biology and Genetics, Izmir Institute of Technology, Gulbahce, 35430 Urla, Izmir, Turkey
| |
Collapse
|
3
|
Monfort T, Azzollini S, Brogard J, Clémençon M, Slembrouck-Brec A, Forster V, Picaud S, Goureau O, Reichman S, Thouvenin O, Grieve K. Dynamic full-field optical coherence tomography module adapted to commercial microscopes allows longitudinal in vitro cell culture study. Commun Biol 2023; 6:992. [PMID: 37770552 PMCID: PMC10539404 DOI: 10.1038/s42003-023-05378-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2023] [Accepted: 09/20/2023] [Indexed: 09/30/2023] Open
Abstract
Dynamic full-field optical coherence tomography (D-FFOCT) has recently emerged as a label-free imaging tool, capable of resolving cell types and organelles within 3D live samples, whilst monitoring their activity at tens of milliseconds resolution. Here, a D-FFOCT module design is presented which can be coupled to a commercial microscope with a stage top incubator, allowing non-invasive label-free longitudinal imaging over periods of minutes to weeks on the same sample. Long term volumetric imaging on human induced pluripotent stem cell-derived retinal organoids is demonstrated, highlighting tissue and cell organization processes such as rosette formation and mitosis as well as cell shape and motility. Imaging on retinal explants highlights single 3D cone and rod structures. An optimal workflow for data acquisition, postprocessing and saving is demonstrated, resulting in a time gain factor of 10 compared to prior state of the art. Finally, a method to increase D-FFOCT signal-to-noise ratio is demonstrated, allowing rapid organoid screening.
Collapse
Affiliation(s)
- Tual Monfort
- Sorbonne Université, INSERM, CNRS, Institut de la Vision, 17 rue Moreau, F-75012, Paris, France
- CHNO des Quinze-Vingts, INSERM-DGOS CIC 1423, 28 rue de Charenton, F-75012, Paris, France
- Paris Eye Imaging Group, Quinze-Vingts National Eye Hospital, INSERM-DGOS, CIC 1423, 28 rue de Charenton, Paris, 75012, France
| | - Salvatore Azzollini
- Sorbonne Université, INSERM, CNRS, Institut de la Vision, 17 rue Moreau, F-75012, Paris, France
| | - Jérémy Brogard
- Sorbonne Université, INSERM, CNRS, Institut de la Vision, 17 rue Moreau, F-75012, Paris, France
| | - Marilou Clémençon
- Sorbonne Université, INSERM, CNRS, Institut de la Vision, 17 rue Moreau, F-75012, Paris, France
| | - Amélie Slembrouck-Brec
- Sorbonne Université, INSERM, CNRS, Institut de la Vision, 17 rue Moreau, F-75012, Paris, France
| | - Valerie Forster
- Sorbonne Université, INSERM, CNRS, Institut de la Vision, 17 rue Moreau, F-75012, Paris, France
| | - Serge Picaud
- Sorbonne Université, INSERM, CNRS, Institut de la Vision, 17 rue Moreau, F-75012, Paris, France
| | - Olivier Goureau
- Sorbonne Université, INSERM, CNRS, Institut de la Vision, 17 rue Moreau, F-75012, Paris, France
| | - Sacha Reichman
- Sorbonne Université, INSERM, CNRS, Institut de la Vision, 17 rue Moreau, F-75012, Paris, France
| | - Olivier Thouvenin
- Institut Langevin, ESPCI Paris, Université PSL, CNRS, 75005, Paris, France
| | - Kate Grieve
- Sorbonne Université, INSERM, CNRS, Institut de la Vision, 17 rue Moreau, F-75012, Paris, France.
- CHNO des Quinze-Vingts, INSERM-DGOS CIC 1423, 28 rue de Charenton, F-75012, Paris, France.
- Paris Eye Imaging Group, Quinze-Vingts National Eye Hospital, INSERM-DGOS, CIC 1423, 28 rue de Charenton, Paris, 75012, France.
| |
Collapse
|
4
|
Phua TJ. Understanding human aging and the fundamental cell signaling link in age-related diseases: the middle-aging hypovascularity hypoxia hypothesis. FRONTIERS IN AGING 2023; 4:1196648. [PMID: 37384143 PMCID: PMC10293850 DOI: 10.3389/fragi.2023.1196648] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 03/30/2023] [Accepted: 05/23/2023] [Indexed: 06/30/2023]
Abstract
Aging-related hypoxia, oxidative stress, and inflammation pathophysiology are closely associated with human age-related carcinogenesis and chronic diseases. However, the connection between hypoxia and hormonal cell signaling pathways is unclear, but such human age-related comorbid diseases do coincide with the middle-aging period of declining sex hormonal signaling. This scoping review evaluates the relevant interdisciplinary evidence to assess the systems biology of function, regulation, and homeostasis in order to discern and decipher the etiology of the connection between hypoxia and hormonal signaling in human age-related comorbid diseases. The hypothesis charts the accumulating evidence to support the development of a hypoxic milieu and oxidative stress-inflammation pathophysiology in middle-aged individuals, as well as the induction of amyloidosis, autophagy, and epithelial-to-mesenchymal transition in aging-related degeneration. Taken together, this new approach and strategy can provide the clarity of concepts and patterns to determine the causes of declining vascularity hemodynamics (blood flow) and physiological oxygenation perfusion (oxygen bioavailability) in relation to oxygen homeostasis and vascularity that cause hypoxia (hypovascularity hypoxia). The middle-aging hypovascularity hypoxia hypothesis could provide the mechanistic interface connecting the endocrine, nitric oxide, and oxygen homeostasis signaling that is closely linked to the progressive conditions of degenerative hypertrophy, atrophy, fibrosis, and neoplasm. An in-depth understanding of these intrinsic biological processes of the developing middle-aged hypoxia could provide potential new strategies for time-dependent therapies in maintaining healthspan for healthy lifestyle aging, medical cost savings, and health system sustainability.
Collapse
Affiliation(s)
- Teow J. Phua
- Molecular Medicine, NSW Health Pathology, John Hunter Hospital, Newcastle, NSW, Australia
| |
Collapse
|
5
|
Mekhileri NV, Major G, Lim K, Mutreja I, Chitcholtan K, Phillips E, Hooper G, Woodfield T. Biofabrication of Modular Spheroids as Tumor-Scale Microenvironments for Drug Screening. Adv Healthc Mater 2023; 12:e2201581. [PMID: 36495232 PMCID: PMC11468982 DOI: 10.1002/adhm.202201581] [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: 06/30/2022] [Revised: 11/13/2022] [Indexed: 12/14/2022]
Abstract
To streamline the drug discovery pipeline, there is a pressing need for preclinical models which replicate the complexity and scale of native tumors. While there have been advancements in the formation of microscale tumor units, these models are cell-line dependent, time-consuming and have not improved clinical trial success rates. In this study, two methods for generating 3D tumor microenvironments are compared, rapidly fabricated hydrogel microspheres and traditional cell-dense spheroids. These modules are then bioassembled into 3D printed thermoplastic scaffolds, using an automated biofabrication process, to form tumor-scale models. Modules are formed with SKOV3 and HFF cells as monocultures and cocultures, and the fabrication efficiency, cell architecture, and drug response profiles are characterized, both as single modules and as multimodular constructs. Cell-encapsulated Gel-MA microspheres are fabricated with high-reproducibility and dimensions necessary for automated tumor-scale bioassembly regardless of cell type, however, only cocultured spheroids form compact modules suitable for bioassembly. Chemosensitivity assays demonstrate the reduced potency of doxorubicin in coculture bioassembled constructs and a ≈five-fold increase in drug resistance of cocultured cells in 3D modules compared with 2D monolayers. This bioassembly system is efficient and tailorable so that a variety of relevant-sized tumor constructs could be developed to study tumorigenesis and modernize drug discovery.
Collapse
Affiliation(s)
- Naveen Vijayan Mekhileri
- Department of Orthopaedic Surgery and Musculoskeletal MedicineCentre for Bioengineering & NanomedicineUniversity of OtagoChristchurchCanterbury8011New Zealand
| | - Gretel Major
- Department of Orthopaedic Surgery and Musculoskeletal MedicineCentre for Bioengineering & NanomedicineUniversity of OtagoChristchurchCanterbury8011New Zealand
| | - Khoon Lim
- Department of Orthopaedic Surgery and Musculoskeletal MedicineCentre for Bioengineering & NanomedicineUniversity of OtagoChristchurchCanterbury8011New Zealand
| | - Isha Mutreja
- Department of Orthopaedic Surgery and Musculoskeletal MedicineCentre for Bioengineering & NanomedicineUniversity of OtagoChristchurchCanterbury8011New Zealand
| | - Kenny Chitcholtan
- Department of Obstetrics and GynaecologyGynaecological Cancer Research GroupUniversity of OtagoChristchurchCanterbury8011New Zealand
| | - Elisabeth Phillips
- Mackenzie Cancer Research GroupDepartment of Pathology and Biomedical ScienceUniversity of OtagoChristchurchCanterbury8011New Zealand
| | - Gary Hooper
- Department of Orthopaedic Surgery and Musculoskeletal MedicineCentre for Bioengineering & NanomedicineUniversity of OtagoChristchurchCanterbury8011New Zealand
| | - Tim Woodfield
- Department of Orthopaedic Surgery and Musculoskeletal MedicineCentre for Bioengineering & NanomedicineUniversity of OtagoChristchurchCanterbury8011New Zealand
| |
Collapse
|
6
|
D'Aiuto N, Hochmann J, Millán M, Di Paolo A, Bologna-Molina R, Sotelo Silveira J, Arocena M. Hypoxia, acidification and oxidative stress in cells cultured at large distances from an oxygen source. Sci Rep 2022; 12:21699. [PMID: 36522457 PMCID: PMC9755289 DOI: 10.1038/s41598-022-26205-y] [Citation(s) in RCA: 23] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2022] [Accepted: 12/12/2022] [Indexed: 12/23/2022] Open
Abstract
Hypoxia is a condition frequently encountered by cells in tissues, whether as a normal feature of their microenvironment or subsequent to deregulated growth. Hypoxia can lead to acidification and increased oxidative stress, with profound consequences for cell physiology and tumorigenesis. Therefore, the interplay between hypoxia and oxidative stress is an important aspect for understanding the effects of hypoxic microenvironments on cells. We have used a previously developed variant of the method of coverslip-induced hypoxia to study the process of acidification in a hypoxic microenvironment and to simultaneously visualize intracellular levels of hypoxia and oxidative stress. We observed high accumulation of CO2 in hypoxic conditions, which we show is the main contributor to acidification in our model. Also, increased levels of oxidative stress were observed in moderately hypoxic cells close to the oxygen source, where the mitochondrial membrane potential was preserved. Conversely, cells at large distances from the oxygen source showed higher levels of hypoxia, milder oxidative stress and reduced mitochondrial membrane potential. Our results contribute to characterize the interplay between reduced oxygen levels, acidification and oxidative stress in a simple in vitro setting, which can be used to model cell responses to an altered environment, such as the early tumor microenvironment.
Collapse
Affiliation(s)
- Natali D'Aiuto
- Cátedra de Bioquímica y Biofísica, Facultad de Odontología, Universidad de la República, Montevideo, Uruguay
- Departamento de Genómica, Instituto de Investigaciones Biológicas Clemente Estable, Montevideo, Uruguay
| | - Jimena Hochmann
- Departamento de Genómica, Instituto de Investigaciones Biológicas Clemente Estable, Montevideo, Uruguay.
- Departamento de Fisiología, Facultad de Medicina, Universidad de la República, Montevideo, Uruguay.
| | - Magdalena Millán
- Departamento de Genómica, Instituto de Investigaciones Biológicas Clemente Estable, Montevideo, Uruguay
| | - Andrés Di Paolo
- Departamento de Genómica, Instituto de Investigaciones Biológicas Clemente Estable, Montevideo, Uruguay
| | - Ronell Bologna-Molina
- Departamento de Patología Molecular, Facultad de Odontología, Universidad de la República, Montevideo, Uruguay
| | - José Sotelo Silveira
- Departamento de Genómica, Instituto de Investigaciones Biológicas Clemente Estable, Montevideo, Uruguay
- Sección Biología Celular, Facultad de Ciencias, Universidad de la República, Montevideo, Uruguay
| | - Miguel Arocena
- Cátedra de Bioquímica y Biofísica, Facultad de Odontología, Universidad de la República, Montevideo, Uruguay.
- Departamento de Genómica, Instituto de Investigaciones Biológicas Clemente Estable, Montevideo, Uruguay.
| |
Collapse
|
7
|
Somasundaram V, Ridnour LA, Cheng RY, Walke AJ, Kedei N, Bhattacharyya DD, Wink AL, Edmondson EF, Butcher D, Warner AC, Dorsey TH, Scheiblin DA, Heinz W, Bryant RJ, Kinders RJ, Lipkowitz S, Wong ST, Pore M, Hewitt SM, McVicar DW, Anderson SK, Chang J, Glynn SA, Ambs S, Lockett SJ, Wink DA. Systemic Nos2 Depletion and Cox inhibition limits TNBC disease progression and alters lymphoid cell spatial orientation and density. Redox Biol 2022; 58:102529. [PMID: 36375380 PMCID: PMC9661390 DOI: 10.1016/j.redox.2022.102529] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2022] [Revised: 10/21/2022] [Accepted: 10/31/2022] [Indexed: 11/10/2022] Open
Abstract
Antitumor immune polarization is a key predictor of clinical outcomes to cancer therapy. An emerging concept influencing clinical outcome involves the spatial location of CD8+ T cells, within the tumor. Our earlier work demonstrated immunosuppressive effects of NOS2 and COX2 tumor expression. Here, we show that NOS2/COX2 levels influence both the polarization and spatial location of lymphoid cells including CD8+ T cells. Importantly, elevated tumor NOS2/COX2 correlated with exclusion of CD8+ T cells from the tumor epithelium. In contrast, tumors expressing low NOS2/COX2 had increased CD8+ T cell penetration into the tumor epithelium. Consistent with a causative relationship between these observations, pharmacological inhibition of COX2 with indomethacin dramatically reduced tumor growth of the 4T1 model of TNBC in both WT and Nos2- mice. This regimen led to complete tumor regression in ∼20-25% of tumor-bearing Nos2- mice, and these animals were resistant to tumor rechallenge. Th1 cytokines were elevated in the blood of treated mice and intratumoral CD4+ and CD8+ T cells were higher in mice that received indomethacin when compared to control untreated mice. Multiplex immunofluorescence imaging confirmed our phenotyping results and demonstrated that targeted Nos2/Cox2 blockade improved CD8+ T cell penetration into the 4T1 tumor core. These findings are consistent with our observations in low NOS2/COX2 expressing breast tumors proving that COX2 activity is responsible for limiting the spatial distribution of effector T cells in TNBC. Together these results suggest that clinically available NSAID's may provide a cost-effective, novel immunotherapeutic approach for treatment of aggressive tumors including triple negative breast cancer.
Collapse
Affiliation(s)
- Veena Somasundaram
- Cancer Innovation Laboratory, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Frederick, MD, USA
| | - Lisa A Ridnour
- Cancer Innovation Laboratory, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Frederick, MD, USA
| | - Robert Ys Cheng
- Cancer Innovation Laboratory, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Frederick, MD, USA
| | - Abigail J Walke
- Optical Microscopy and Analysis Laboratory, Frederick National Laboratory for Cancer Research, Leidos Biomedical Research Inc. for the National Cancer Institute, Frederick, MD, USA
| | - Noemi Kedei
- Collaborative Protein Technology Resource Nanoscale Protein Analysis, Office of Science Technology Resources, CCR, NCI, NIH, Bethesda, MD, USA
| | - Dibyangana D Bhattacharyya
- Cancer Innovation Laboratory, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Frederick, MD, USA
| | - Adelaide L Wink
- Optical Microscopy and Analysis Laboratory, Frederick National Laboratory for Cancer Research, Leidos Biomedical Research Inc. for the National Cancer Institute, Frederick, MD, USA
| | - Elijah F Edmondson
- Molecular Histopathology Laboratories, Leidos Biomedical Research Inc. for NCI, Frederick, MD, USA
| | - Donna Butcher
- Molecular Histopathology Laboratories, Leidos Biomedical Research Inc. for NCI, Frederick, MD, USA
| | - Andrew C Warner
- Molecular Histopathology Laboratories, Leidos Biomedical Research Inc. for NCI, Frederick, MD, USA
| | - Tiffany H Dorsey
- Laboratory of Human Carcinogenesis, CCR, NCI, NIH, Bethesda, MD, USA
| | - David A Scheiblin
- Optical Microscopy and Analysis Laboratory, Frederick National Laboratory for Cancer Research, Leidos Biomedical Research Inc. for the National Cancer Institute, Frederick, MD, USA
| | - William Heinz
- Optical Microscopy and Analysis Laboratory, Frederick National Laboratory for Cancer Research, Leidos Biomedical Research Inc. for the National Cancer Institute, Frederick, MD, USA
| | - Richard J Bryant
- Nuffield Department of Surgical Sciences, University of Oxford, Oxford, UK
| | - Robert J Kinders
- Office of the Director, Division of Cancer Treatment and Diagnosis, NCI, Frederick, MD, USA
| | | | - Stephen Tc Wong
- Systems Medicine and Bioengineering, Houston Methodist Neal Cancer Center and Weill Cornell Medical College, Houston, TX, USA
| | - Milind Pore
- Imaging Mass Cytometry Frederick National Laboratory for Cancer Research, USA
| | | | - Daniel W McVicar
- Cancer Innovation Laboratory, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Frederick, MD, USA
| | - Stephen K Anderson
- Cancer Innovation Laboratory, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Frederick, MD, USA; Basic Science Program, Frederick National Laboratory for Cancer Research, Frederick, MD, USA
| | - Jenny Chang
- Mary and Ron Neal Cancer Center, Houston Methodist Weill Cornell Medical College, Houston, TX, USA
| | - Sharon A Glynn
- Discipline of Pathology, Lambe Institute for Translational Research, School of Medicine, University of Galway, Galway, H91 TK33, Ireland
| | - Stefan Ambs
- Laboratory of Human Carcinogenesis, CCR, NCI, NIH, Bethesda, MD, USA
| | - Stephen J Lockett
- Optical Microscopy and Analysis Laboratory, Frederick National Laboratory for Cancer Research, Leidos Biomedical Research Inc. for the National Cancer Institute, Frederick, MD, USA.
| | - David A Wink
- Cancer Innovation Laboratory, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Frederick, MD, USA.
| |
Collapse
|
8
|
Cellular Model of Malignant Transformation of Primary Human Astrocytes Induced by Deadhesion/Readhesion Cycles. Int J Mol Sci 2022; 23:ijms23094471. [PMID: 35562862 PMCID: PMC9103552 DOI: 10.3390/ijms23094471] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2021] [Revised: 04/03/2022] [Accepted: 04/12/2022] [Indexed: 01/07/2023] Open
Abstract
Astrocytoma is the most common and aggressive tumor of the central nervous system. Genetic and environmental factors, bacterial infection, and several other factors are known to be involved in gliomagenesis, although the complete underlying molecular mechanism is not fully understood. Tumorigenesis is a multistep process involving initiation, promotion, and progression. We present a human model of malignant astrocyte transformation established by subjecting primary astrocytes from healthy adults to four sequential cycles of forced anchorage impediment (deadhesion). After limiting dilution of the surviving cells obtained after the fourth deadhesion/readhesion cycle, three clones were randomly selected, and exhibited malignant characteristics, including increased proliferation rate and capacity for colony formation, migration, and anchorage-independent growth in soft agar. Functional assay results for these clonal cells, including response to temozolomide, were comparable to U87MG—a human glioblastoma-derived cell lineage—reinforcing malignant cell transformation. RNA-Seq analysis by next-generation sequencing of the transformed clones relative to the primary astrocytes revealed upregulation of genes involved in the PI3K/AKT and Wnt/β-catenin signaling pathways, in addition to upregulation of genes related to epithelial–mesenchymal transition, and downregulation of genes related to aerobic respiration. These findings, at a molecular level, corroborate the change in cell behavior towards mesenchymal-like cell dedifferentiation. This linear progressive model of malignant human astrocyte transformation is unique in that neither genetic manipulation nor treatment with carcinogens are used, representing a promising tool for testing combined therapeutic strategies for glioblastoma patients, and furthering knowledge of astrocytoma transformation and progression.
Collapse
|
9
|
Mukundan S, Singh P, Shah A, Kumar R, O’Neill KC, Carter CL, Russell DG, Subbian S, Parekkadan B. In Vitro Miniaturized Tuberculosis Spheroid Model. Biomedicines 2021; 9:1209. [PMID: 34572395 PMCID: PMC8470281 DOI: 10.3390/biomedicines9091209] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2021] [Revised: 09/01/2021] [Accepted: 09/06/2021] [Indexed: 12/11/2022] Open
Abstract
Tuberculosis (TB) is a public health concern that impacts 10 million people around the world. Current in vitro models are low throughput and/or lack caseation, which impairs drug effectiveness in humans. Here, we report the generation of THP-1 human monocyte/macrophage spheroids housing mycobacteria (TB spheroids). These TB spheroids have a central core of dead cells co-localized with mycobacteria and are hypoxic. TB spheroids exhibit higher levels of pro-inflammatory factor TNFα and growth factors G-CSF and VEGF when compared to non-infected control. TB spheroids show high levels of lipid deposition, characterized by MALDI mass spectrometry imaging. TB spheroids infected with strains of differential virulence, Mycobacterium tuberculosis (Mtb) HN878 and CDC1551 vary in response to Isoniazid and Rifampicin. Finally, we adapt the spheroid model to form peripheral blood mononuclear cells (PBMCs) and lung fibroblasts (NHLF) 3D co-cultures. These results pave the way for the development of new strategies for disease modeling and therapeutic discovery.
Collapse
Affiliation(s)
- Shilpaa Mukundan
- Department of Biomedical Engineering, Rutgers, The State University of New Jersey, Jersey City, NJ 08854, USA; (S.M.); (A.S.)
| | - Pooja Singh
- Public Health Research Institute, New Jersey Medical School, Rutgers, The State University of New Jersey, Jersey City, NJ 07103, USA; (P.S.); (R.K.); (S.S.)
| | - Aditi Shah
- Department of Biomedical Engineering, Rutgers, The State University of New Jersey, Jersey City, NJ 08854, USA; (S.M.); (A.S.)
| | - Ranjeet Kumar
- Public Health Research Institute, New Jersey Medical School, Rutgers, The State University of New Jersey, Jersey City, NJ 07103, USA; (P.S.); (R.K.); (S.S.)
| | - Kelly C. O’Neill
- Department Center for Discovery and Innovation, Hackensack Meridian Health, Neptune, NJ 07110, USA; (K.C.O.); (C.L.C.)
| | - Claire L. Carter
- Department Center for Discovery and Innovation, Hackensack Meridian Health, Neptune, NJ 07110, USA; (K.C.O.); (C.L.C.)
| | - David G. Russell
- Department of Microbiology and Immunology, School of Veterinary Medicine, Cornell University, Ithaca, NY 14853, USA;
| | - Selvakumar Subbian
- Public Health Research Institute, New Jersey Medical School, Rutgers, The State University of New Jersey, Jersey City, NJ 07103, USA; (P.S.); (R.K.); (S.S.)
| | - Biju Parekkadan
- Department of Biomedical Engineering, Rutgers, The State University of New Jersey, Jersey City, NJ 08854, USA; (S.M.); (A.S.)
- Department of Medicine, Rutgers Biomedical Health Sciences, Rutgers, The State University of New Jersey, Jersey City, NJ 08854, USA
| |
Collapse
|