1
|
Tavakoli N, Fong EJ, Coleman A, Huang YK, Bigger M, Doche ME, Kim S, Lenz HJ, Graham NA, Macklin P, Finley SD, Mumenthaler SM. Merging Metabolic Modeling and Imaging for Screening Therapeutic Targets in Colorectal Cancer. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.05.24.595756. [PMID: 38826317 PMCID: PMC11142224 DOI: 10.1101/2024.05.24.595756] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/04/2024]
Abstract
Cancer-associated fibroblasts (CAFs) play a key role in metabolic reprogramming and are well-established contributors to drug resistance in colorectal cancer (CRC). To exploit this metabolic crosstalk, we integrated a systems biology approach that identified key metabolic targets in a data-driven method and validated them experimentally. This process involved a novel machine learning-based method to computationally screen, in a high-throughput manner, the effects of enzyme perturbations predicted by a computational model of CRC metabolism. This approach reveals the network-wide effects of metabolic perturbations. Our results highlighted hexokinase (HK) as the crucial target, which subsequently became our focus for experimental validation using patient-derived tumor organoids (PDTOs). Through metabolic imaging and viability assays, we found that PDTOs cultured in CAF-conditioned media exhibited increased sensitivity to HK inhibition, confirming the model predictions. Our approach emphasizes the critical role of integrating computational and experimental techniques in exploring and exploiting CRC-CAF crosstalk.
Collapse
Affiliation(s)
- Niki Tavakoli
- Alfred E. Mann Department of Biomedical Engineering, University of Southern California, Los Angeles, CA, 90089, USA
| | - Emma J. Fong
- Ellison Institute of Technology, Los Angeles, CA, 90064, USA
| | - Abigail Coleman
- Ellison Institute of Technology, Los Angeles, CA, 90064, USA
| | - Yu-Kai Huang
- Ellison Institute of Technology, Los Angeles, CA, 90064, USA
| | - Mathias Bigger
- Ellison Institute of Technology, Los Angeles, CA, 90064, USA
- Mork Family Department of Chemical Engineering and Materials Science, University of Southern California, Los Angeles, CA, 90089, USA
| | | | - Seungil Kim
- Ellison Institute of Technology, Los Angeles, CA, 90064, USA
| | - Heinz-Josef Lenz
- Division of Medical Oncology, Norris Comprehensive Cancer Center, University of Southern California, Los Angeles, CA, 90033, USA
| | - Nicholas A. Graham
- Mork Family Department of Chemical Engineering and Materials Science, University of Southern California, Los Angeles, CA, 90089, USA
| | - Paul Macklin
- Department of Intelligent Systems Engineering, Indiana University, Bloomington, IN, 46202, USA
| | - Stacey D. Finley
- Alfred E. Mann Department of Biomedical Engineering, University of Southern California, Los Angeles, CA, 90089, USA
- Mork Family Department of Chemical Engineering and Materials Science, University of Southern California, Los Angeles, CA, 90089, USA
- Department of Quantitative and Computational Biology, University of Southern California, Los Angeles, CA, 90089, USA
| | - Shannon M. Mumenthaler
- Alfred E. Mann Department of Biomedical Engineering, University of Southern California, Los Angeles, CA, 90089, USA
- Ellison Institute of Technology, Los Angeles, CA, 90064, USA
- Division of Medical Oncology, Norris Comprehensive Cancer Center, University of Southern California, Los Angeles, CA, 90033, USA
| |
Collapse
|
2
|
Tedeschi G, Palomba F, Scipioni L, Digman MA. Multimodal Phasor Approach to study breast cancer cells invasion in 3D spheroid model. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.06.10.598307. [PMID: 38915530 PMCID: PMC11195137 DOI: 10.1101/2024.06.10.598307] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/26/2024]
Abstract
We implemented a multimodal set of functional imaging techniques optimized for deep-tissue imaging to investigate how cancer cells invade surrounding tissues and how their physiological properties change in the process. As a model for cancer invasion of the extracellular matrix, we created 3D spheroids from triple-negative breast cancer cells (MDA-MB-231) and non-tumorigenic breast epithelial cells (MCF-10A). We analyzed multiple hallmarks of cancer within the same spheroid by combining a number of imaging techniques, such as metabolic imaging of NADH by Fluorescence Lifetime Imaging Microscopy (NADH-FLIM), hyperspectral imaging of a solvatochromic lipophilic dye (Nile Red) and extracellular matrix imaging by Second Harmonic Generation (SHG). We included phasor-based bioimage analysis of spheroids at three different time points, tracking both morphological and biological properties, including cellular metabolism, fatty acids storage, and collagen organization. Employing this multimodal deep-imaging framework, we observed and quantified cancer cell plasticity in response to changes in the environment composition.
Collapse
Affiliation(s)
- Giulia Tedeschi
- Laboratory for Fluorescence Dynamics, Biomedical Engineering Department, University of California, Irvine, Irvine, CA 92617 (USA)
| | - Francesco Palomba
- Laboratory for Fluorescence Dynamics, Biomedical Engineering Department, University of California, Irvine, Irvine, CA 92617 (USA)
| | - Lorenzo Scipioni
- Laboratory for Fluorescence Dynamics, Biomedical Engineering Department, University of California, Irvine, Irvine, CA 92617 (USA)
| | - Michelle A Digman
- Laboratory for Fluorescence Dynamics, Biomedical Engineering Department, University of California, Irvine, Irvine, CA 92617 (USA)
| |
Collapse
|
3
|
Pickett MR, Chen YI, Kamra M, Kumar S, Kalkunte N, Sugerman GP, Varodom K, Rausch MK, Zoldan J, Yeh HC, Parekh SH. Assessing the impact of extracellular matrix fiber orientation on breast cancer cellular metabolism. Cancer Cell Int 2024; 24:199. [PMID: 38840117 PMCID: PMC11151503 DOI: 10.1186/s12935-024-03385-3] [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: 11/16/2023] [Accepted: 05/25/2024] [Indexed: 06/07/2024] Open
Abstract
The extracellular matrix (ECM) is a dynamic and complex microenvironment that modulates cell behavior and cell fate. Changes in ECM composition and architecture have been correlated with development, differentiation, and disease progression in various pathologies, including breast cancer [1]. Studies have shown that aligned fibers drive a pro-metastatic microenvironment, promoting the transformation of mammary epithelial cells into invasive ductal carcinoma via the epithelial-to-mesenchymal transition (EMT) [2]. The impact of ECM orientation on breast cancer metabolism, however, is largely unknown. Here, we employ two non-invasive imaging techniques, fluorescence-lifetime imaging microscopy (FLIM) and intensity-based multiphoton microscopy, to assess the metabolic states of cancer cells cultured on ECM-mimicking nanofibers in a random and aligned orientation. By tracking the changes in the intrinsic fluorescence of nicotinamide adenine dinucleotide and flavin adenine dinucleotide, as well as expression levels of metastatic markers, we reveal how ECM fiber orientation alters cancer metabolism and EMT progression. Our study indicates that aligned cellular microenvironments play a key role in promoting metastatic phenotypes of breast cancer as evidenced by a more glycolytic metabolic signature on nanofiber scaffolds of aligned orientation compared to scaffolds of random orientation. This finding is particularly relevant for subsets of breast cancer marked by high levels of collagen remodeling (e.g. pregnancy associated breast cancer), and may serve as a platform for predicting clinical outcomes within these subsets [3-6].
Collapse
Affiliation(s)
- Madison R Pickett
- Department of Biomedical Engineering, The University of Texas at Austin, 107 W Dean Keeton Street Stop C0800, Austin, TX, 78712, USA.
| | - Yuan-I Chen
- Department of Biomedical Engineering, The University of Texas at Austin, 107 W Dean Keeton Street Stop C0800, Austin, TX, 78712, USA
| | - Mohini Kamra
- Department of Biomedical Engineering, The University of Texas at Austin, 107 W Dean Keeton Street Stop C0800, Austin, TX, 78712, USA
| | - Sachin Kumar
- Department of Biomedical Engineering, The University of Texas at Austin, 107 W Dean Keeton Street Stop C0800, Austin, TX, 78712, USA
- Centre for Biomedical Engineering, Indian Institute of Technology Delhi, New Delhi, 110016, India
| | - Nikhith Kalkunte
- Department of Biomedical Engineering, The University of Texas at Austin, 107 W Dean Keeton Street Stop C0800, Austin, TX, 78712, USA
| | - Gabriella P Sugerman
- Department of Biomedical Engineering, The University of Texas at Austin, 107 W Dean Keeton Street Stop C0800, Austin, TX, 78712, USA
| | - Kelsey Varodom
- Department of Biomedical Engineering, The University of Texas at Austin, 107 W Dean Keeton Street Stop C0800, Austin, TX, 78712, USA
| | - Manuel K Rausch
- Department of Biomedical Engineering, The University of Texas at Austin, 107 W Dean Keeton Street Stop C0800, Austin, TX, 78712, USA
- Department of Aerospace Engineering and Engineering Mechanics, The University of Texas at Austin, 78712, Austin, TX, USA
- Department of Mechanical Engineering, The University of Texas at Austin, 78712, Austin, TX, USA
- Oden Institute for Computational Engineering and Sciences, The University of Texas at Austin, 78712, Austin, TX, USA
| | - Janet Zoldan
- Department of Biomedical Engineering, The University of Texas at Austin, 107 W Dean Keeton Street Stop C0800, Austin, TX, 78712, USA
| | - Hsin-Chin Yeh
- Department of Biomedical Engineering, The University of Texas at Austin, 107 W Dean Keeton Street Stop C0800, Austin, TX, 78712, USA
- Texas Materials Institute, The University of Texas at Austin, Austin, TX, USA
| | - Sapun H Parekh
- Department of Biomedical Engineering, The University of Texas at Austin, 107 W Dean Keeton Street Stop C0800, Austin, TX, 78712, USA.
| |
Collapse
|
4
|
Liu Y, Wang YJ, Du Y, Liu W, Huang X, Fan Z, Lu J, Yi R, Xiang XW, Xia X, Gu H, Liu YJ, Liu B. DNA nanomachines reveal an adaptive energy mode in confinement-induced amoeboid migration powered by polarized mitochondrial distribution. Proc Natl Acad Sci U S A 2024; 121:e2317492121. [PMID: 38547056 PMCID: PMC10998588 DOI: 10.1073/pnas.2317492121] [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: 10/10/2023] [Accepted: 02/19/2024] [Indexed: 04/02/2024] Open
Abstract
Energy metabolism is highly interdependent with adaptive cell migration in vivo. Mechanical confinement is a critical physical cue that induces switchable migration modes of the mesenchymal-to-amoeboid transition (MAT). However, the energy states in distinct migration modes, especially amoeboid-like stable bleb (A2) movement, remain unclear. In this report, we developed multivalent DNA framework-based nanomachines to explore strategical mitochondrial trafficking and differential ATP levels during cell migration in mechanically heterogeneous microenvironments. Through single-particle tracking and metabolomic analysis, we revealed that fast A2-moving cells driven by biomimetic confinement recruited back-end positioning of mitochondria for powering highly polarized cytoskeletal networks, preferentially adopting an energy-saving mode compared with a mesenchymal mode of cell migration. We present a versatile DNA nanotool for cellular energy exploration and highlight that adaptive energy strategies coordinately support switchable migration modes for facilitating efficient metastatic escape, offering a unique perspective for therapeutic interventions in cancer metastasis.
Collapse
Affiliation(s)
- Yixin Liu
- Department of Chemistry, Shanghai Stomatological Hospital, State Key Lab of Molecular Engineering of Polymers, Shanghai Xuhui Central Hospital, Zhongshan-Xuhui Hospital, Shanghai Key Laboratory of Medical Epigenetics, Institutes of Biomedical Sciences, Fudan University, Shanghai200438, China
| | - Ya-Jun Wang
- Department of Chemistry, Shanghai Stomatological Hospital, State Key Lab of Molecular Engineering of Polymers, Shanghai Xuhui Central Hospital, Zhongshan-Xuhui Hospital, Shanghai Key Laboratory of Medical Epigenetics, Institutes of Biomedical Sciences, Fudan University, Shanghai200438, China
| | - Yang Du
- Department of Chemistry, Shanghai Stomatological Hospital, State Key Lab of Molecular Engineering of Polymers, Shanghai Xuhui Central Hospital, Zhongshan-Xuhui Hospital, Shanghai Key Laboratory of Medical Epigenetics, Institutes of Biomedical Sciences, Fudan University, Shanghai200438, China
| | - Wei Liu
- Department of Chemistry, Shanghai Stomatological Hospital, State Key Lab of Molecular Engineering of Polymers, Shanghai Xuhui Central Hospital, Zhongshan-Xuhui Hospital, Shanghai Key Laboratory of Medical Epigenetics, Institutes of Biomedical Sciences, Fudan University, Shanghai200438, China
| | - Xuedong Huang
- Department of Chemistry, Shanghai Stomatological Hospital, State Key Lab of Molecular Engineering of Polymers, Shanghai Xuhui Central Hospital, Zhongshan-Xuhui Hospital, Shanghai Key Laboratory of Medical Epigenetics, Institutes of Biomedical Sciences, Fudan University, Shanghai200438, China
| | - Zihui Fan
- Department of Chemistry, Shanghai Stomatological Hospital, State Key Lab of Molecular Engineering of Polymers, Shanghai Xuhui Central Hospital, Zhongshan-Xuhui Hospital, Shanghai Key Laboratory of Medical Epigenetics, Institutes of Biomedical Sciences, Fudan University, Shanghai200438, China
| | - Jiayin Lu
- Department of Chemistry, Shanghai Stomatological Hospital, State Key Lab of Molecular Engineering of Polymers, Shanghai Xuhui Central Hospital, Zhongshan-Xuhui Hospital, Shanghai Key Laboratory of Medical Epigenetics, Institutes of Biomedical Sciences, Fudan University, Shanghai200438, China
| | - Runqiu Yi
- Department of Chemistry, Shanghai Stomatological Hospital, State Key Lab of Molecular Engineering of Polymers, Shanghai Xuhui Central Hospital, Zhongshan-Xuhui Hospital, Shanghai Key Laboratory of Medical Epigenetics, Institutes of Biomedical Sciences, Fudan University, Shanghai200438, China
| | - Xiao-Wei Xiang
- Department of Chemistry, Shanghai Stomatological Hospital, State Key Lab of Molecular Engineering of Polymers, Shanghai Xuhui Central Hospital, Zhongshan-Xuhui Hospital, Shanghai Key Laboratory of Medical Epigenetics, Institutes of Biomedical Sciences, Fudan University, Shanghai200438, China
| | - Xinwei Xia
- Department of Chemistry, Shanghai Stomatological Hospital, State Key Lab of Molecular Engineering of Polymers, Shanghai Xuhui Central Hospital, Zhongshan-Xuhui Hospital, Shanghai Key Laboratory of Medical Epigenetics, Institutes of Biomedical Sciences, Fudan University, Shanghai200438, China
| | - Hongzhou Gu
- Department of Chemistry, Shanghai Stomatological Hospital, State Key Lab of Molecular Engineering of Polymers, Shanghai Xuhui Central Hospital, Zhongshan-Xuhui Hospital, Shanghai Key Laboratory of Medical Epigenetics, Institutes of Biomedical Sciences, Fudan University, Shanghai200438, China
| | - Yan-Jun Liu
- Department of Chemistry, Shanghai Stomatological Hospital, State Key Lab of Molecular Engineering of Polymers, Shanghai Xuhui Central Hospital, Zhongshan-Xuhui Hospital, Shanghai Key Laboratory of Medical Epigenetics, Institutes of Biomedical Sciences, Fudan University, Shanghai200438, China
| | - Baohong Liu
- Department of Chemistry, Shanghai Stomatological Hospital, State Key Lab of Molecular Engineering of Polymers, Shanghai Xuhui Central Hospital, Zhongshan-Xuhui Hospital, Shanghai Key Laboratory of Medical Epigenetics, Institutes of Biomedical Sciences, Fudan University, Shanghai200438, China
| |
Collapse
|
5
|
Passos J, Lopes LB, Panitch A. Collagen-Binding Nanoparticles for Paclitaxel Encapsulation and Breast Cancer Treatment. ACS Biomater Sci Eng 2023; 9:6805-6820. [PMID: 37982792 PMCID: PMC10716849 DOI: 10.1021/acsbiomaterials.3c01332] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2023] [Revised: 10/28/2023] [Accepted: 10/30/2023] [Indexed: 11/21/2023]
Abstract
In this study, we developed a novel hybrid collagen-binding nanocarrier for potential intraductal administration and local breast cancer treatment. The particles were formed by the encapsulation of nanostructured lipid carriers (NLCs) containing the cytotoxic drug paclitaxel within a shell of poly(N-isopropylacrylamide) (pNIPAM), and were functionalized with SILY, a peptide that binds to collagen type I (which is overexpressed in the mammary tumor microenvironment) to improve local retention and selectivity. The encapsulation of the NLCs in the pNIPAM shell increased nanoparticle size by approximately 140 nm, and after purification, a homogeneous system of hybrid nanoparticles (∼96%) was obtained. The nanoparticles exhibited high loading efficiency (<76%) and were capable of prolonging paclitaxel release for up to 120 h. SILY-modified nanoparticles showed the ability to bind to collagen-coated surfaces and naturally elaborated collagen. Hybrid nanoparticles presented cytotoxicity up to 3.7-fold higher than pNIPAM-only nanoparticles on mammary tumor cells cultured in monolayers. In spheroids, the increase in cytotoxicity was up to 1.8-fold. Compared to lipid nanoparticles, the hybrid nanoparticle modified with SILY increased the viability of nontumor breast cells by up to 1.59-fold in a coculture model, suggesting the effectiveness and safety of the system.
Collapse
Affiliation(s)
- Julia
Sapienza Passos
- Wallace
H. Coulter Department of Biomedical Engineering, Georgia Institute of Technology and Emory University, Atlanta, Georgia 30332, United States
- Department
of Pharmacology, Institute of Biomedical Sciences, University of Sao Paulo, Sao Paulo, SP 05508-000, Brazil
| | - Luciana B. Lopes
- Department
of Pharmacology, Institute of Biomedical Sciences, University of Sao Paulo, Sao Paulo, SP 05508-000, Brazil
| | - Alyssa Panitch
- Wallace
H. Coulter Department of Biomedical Engineering, Georgia Institute of Technology and Emory University, Atlanta, Georgia 30332, United States
| |
Collapse
|
6
|
Poonja S, Forero Pinto A, Lloyd MC, Damaghi M, Rejniak KA. Dynamics of Fibril Collagen Remodeling by Tumor Cells: A Model of Tumor-Associated Collagen Signatures. Cells 2023; 12:2688. [PMID: 38067116 PMCID: PMC10705683 DOI: 10.3390/cells12232688] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2023] [Revised: 11/01/2023] [Accepted: 11/17/2023] [Indexed: 12/18/2023] Open
Abstract
Many solid tumors are characterized by a dense extracellular matrix (ECM) composed of various ECM fibril proteins. These proteins provide structural support and a biological context for the residing cells. The reciprocal interactions between growing and migrating tumor cells and the surrounding stroma result in dynamic changes in the ECM architecture and its properties. With the use of advanced imaging techniques, several specific patterns in the collagen surrounding the breast tumor have been identified in both tumor murine models and clinical histology images. These tumor-associated collagen signatures (TACS) include loosely organized fibrils far from the tumor and fibrils aligned either parallel or perpendicular to tumor colonies. They are correlated with tumor behavior, such as benign growth or invasive migration. However, it is not fully understood how one specific fibril pattern can be dynamically remodeled to form another alignment. Here, we present a novel multi-cellular lattice-free (MultiCell-LF) agent-based model of ECM that, in contrast to static histology images, can simulate dynamic changes between TACSs. This model allowed us to identify the rules of cell-ECM physical interplay and feedback that guided the emergence and transition among various TACSs.
Collapse
Affiliation(s)
- Sharan Poonja
- Integrated Mathematical Oncology Department, H. Lee Moffitt Cancer Center, Research Institute, Tampa, FL 33612, USA
| | - Ana Forero Pinto
- Integrated Mathematical Oncology Department, H. Lee Moffitt Cancer Center, Research Institute, Tampa, FL 33612, USA
- Cancer Biology PhD Program, University of South Florida, Tampa, FL 33612, USA
| | - Mark C. Lloyd
- Fujifilm Healthcare US, Inc., Lexington, MA 02421, USA;
| | - Mehdi Damaghi
- Department of Pathology, Renaissance School of Medicine, Stony Brook University, Stony Brook, NY 11794, USA
| | - Katarzyna A. Rejniak
- Integrated Mathematical Oncology Department, H. Lee Moffitt Cancer Center, Research Institute, Tampa, FL 33612, USA
- Department of Oncologic Sciences, Morsani School of Medicine, University of South Florida, Tampa, FL 33612, USA
| |
Collapse
|
7
|
Sohrabi A, Lefebvre AEYT, Harrison MJ, Condro MC, Sanazzaro TM, Safarians G, Solomon I, Bastola S, Kordbacheh S, Toh N, Kornblum HI, Digman MA, Seidlits SK. Microenvironmental stiffness induces metabolic reprogramming in glioblastoma. Cell Rep 2023; 42:113175. [PMID: 37756163 PMCID: PMC10842372 DOI: 10.1016/j.celrep.2023.113175] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2023] [Revised: 08/28/2023] [Accepted: 09/07/2023] [Indexed: 09/29/2023] Open
Abstract
The mechanical properties of solid tumors influence tumor cell phenotype and the ability to invade surrounding tissues. Using bioengineered scaffolds to provide a matrix microenvironment for patient-derived glioblastoma (GBM) spheroids, this study demonstrates that a soft, brain-like matrix induces GBM cells to shift to a glycolysis-weighted metabolic state, which supports invasive behavior. We first show that orthotopic murine GBM tumors are stiffer than peritumoral brain tissues, but tumor stiffness is heterogeneous where tumor edges are softer than the tumor core. We then developed 3D scaffolds with μ-compressive moduli resembling either stiffer tumor core or softer peritumoral brain tissue. We demonstrate that the softer matrix microenvironment induces a shift in GBM cell metabolism toward glycolysis, which manifests in lower proliferation rate and increased migration activities. Finally, we show that these mechanical cues are transduced from the matrix via CD44 and integrin receptors to induce metabolic and phenotypic changes in cancer cells.
Collapse
Affiliation(s)
- Alireza Sohrabi
- Department of Biomedical Engineering, University of Texas at Austin, Austin, TX 78712, USA; Department of Bioengineering, University of California, Los Angeles, Los Angeles, CA 90095, USA
| | - Austin E Y T Lefebvre
- Department of Biomedical Engineering, University of California at Irvine, Irvine, CA 92697, USA
| | - Mollie J Harrison
- Department of Biomedical Engineering, University of Texas at Austin, Austin, TX 78712, USA
| | - Michael C Condro
- Department of Molecular and Medical Pharmacology, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA 90095, USA
| | - Talia M Sanazzaro
- Department of Biomedical Engineering, University of Texas at Austin, Austin, TX 78712, USA
| | - Gevick Safarians
- Department of Bioengineering, University of California, Los Angeles, Los Angeles, CA 90095, USA
| | - Itay Solomon
- Department of Bioengineering, University of California, Los Angeles, Los Angeles, CA 90095, USA
| | - Soniya Bastola
- Department of Molecular and Medical Pharmacology, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA 90095, USA
| | - Shadi Kordbacheh
- Department of Bioengineering, University of California, Los Angeles, Los Angeles, CA 90095, USA
| | - Nadia Toh
- Department of Biomedical Engineering, University of Texas at Austin, Austin, TX 78712, USA
| | - Harley I Kornblum
- Department of Molecular and Medical Pharmacology, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA 90095, USA
| | - Michelle A Digman
- Department of Biomedical Engineering, University of California at Irvine, Irvine, CA 92697, USA
| | - Stephanie K Seidlits
- Department of Biomedical Engineering, University of Texas at Austin, Austin, TX 78712, USA; Department of Bioengineering, University of California, Los Angeles, Los Angeles, CA 90095, USA.
| |
Collapse
|
8
|
Bertolio R, Napoletano F, Del Sal G. Dynamic links between mechanical forces and metabolism shape the tumor milieu. Curr Opin Cell Biol 2023; 84:102218. [PMID: 37597464 DOI: 10.1016/j.ceb.2023.102218] [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: 06/17/2023] [Revised: 07/19/2023] [Accepted: 07/20/2023] [Indexed: 08/21/2023]
Abstract
Cell function relies on the spatiotemporal dynamics of metabolic reactions. In all physiopathological processes of tissues, mechanical forces impact the structure and function of membranes, enzymes, organelles and regulators of metabolic gene programs, thus regulating cell metabolism. In turn, metabolic pathways feedback impacts the physical properties of cell and tissues. Hence, metabolism and tissue mechanics are dynamically intertwined and continuously interact. Cancer is akin to an ecosystem, comprising tumor cells and various subpopulations of stromal cells embedded in an altered extracellular matrix. The progression of cancer, from initiation to advanced stage and metastasis, is driven by genetic mutations and crucially influenced by physical and metabolic alterations in the tumor microenvironment. These alterations also play a pivotal role in cancer cells evasion from immune surveillance and in developing resistance to treatments. Here, we highlight emerging evidence showing that mechano-metabolic circuits in cancer and stromal cells regulate multiple processes crucial for tumor progression and discuss potential approaches to improve therapeutic treatments by interfering with these circuits.
Collapse
Affiliation(s)
- Rebecca Bertolio
- Department of Life Sciences, University of Trieste, 34127 Trieste, Italy; International Centre for Genetic Engineering and Biotechnology (ICGEB), Area Science Park-Padriciano, 34149 Trieste, Italy
| | - Francesco Napoletano
- Department of Life Sciences, University of Trieste, 34127 Trieste, Italy; International Centre for Genetic Engineering and Biotechnology (ICGEB), Area Science Park-Padriciano, 34149 Trieste, Italy
| | - Giannino Del Sal
- Department of Life Sciences, University of Trieste, 34127 Trieste, Italy; International Centre for Genetic Engineering and Biotechnology (ICGEB), Area Science Park-Padriciano, 34149 Trieste, Italy; IFOM ETS, The AIRC Institute of Molecular Oncology, Milan, Italy.
| |
Collapse
|
9
|
Nair L, Mukherjee S, Kaur K, Murphy CM, Ravichandiran V, Roy S, Singh M. Multi compartmental 3D breast cancer disease model–recapitulating tumor complexity in in-vitro. Biochim Biophys Acta Gen Subj 2023; 1867:130361. [PMID: 37019341 DOI: 10.1016/j.bbagen.2023.130361] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2023] [Revised: 03/26/2023] [Accepted: 03/29/2023] [Indexed: 04/05/2023]
Abstract
Breast cancer is the most common ailment among women. In 2020, it had the highest incidence of any type of cancer. Many Phase II and III anti-cancer drugs fail due to efficacy, durability, and side effects. Thus, accelerated drug screening models must be accurate. In-vivo models have been used for a long time, but delays, inconsistent results, and a greater sense of responsibility among scientists toward wildlife have led to the search for in-vitro alternatives. Stromal components support breast cancer growth and survival. Multi-compartment Transwell models may be handy instruments. Co-culturing breast cancer cells with endothelium and fibroblasts improves modelling. The extracellular matrix (ECM) supports native 3D hydrogels in natural and polymeric forms. 3D Transwell cultured tumor spheroids mimicked in-vivo pathological conditions. Tumor invasion, migration, Trans-endothelial migration, angiogenesis, and spread are studied using comprehensive models. Transwell models can create a cancer niche and conduct high-throughput drug screening, promising future applications. Our comprehensive shows how 3D in-vitro multi compartmental models may be useful in producing breast cancer stroma in Transwell culture.
Collapse
Affiliation(s)
- Lakshmi Nair
- Department of Pharmaceutical Sciences, Assam Central University, Silchar, Assam 788011, India
| | - Souvik Mukherjee
- Department of Pharmaceutical Sciences, Guru Ghasidas University, Koni, Bilaspur,(C.G 495009, India
| | - Kulwinder Kaur
- Tissue Engineering Research Group, Department of Anatomy & Regenerative Medicine, Royal College of Surgeons (RCSI), Dublin D02YN77, Ireland
| | - Ciara M Murphy
- Tissue Engineering Research Group, Department of Anatomy & Regenerative Medicine, Royal College of Surgeons (RCSI), Dublin D02YN77, Ireland; Trinity Centre for Biomedical Engineering, Trinity College Dublin (TCD), Dublin D02YN77, Ireland; Advanced Materials and Bioengineering Research Centre (AMBER), RCSI and TCD, Dublin, Ireland
| | - Velayutham Ravichandiran
- Department of Pharmacology and Toxicology, National Institute of Pharmaceutical Education and Research, Kolkata, West Bengal 700054, India
| | - Subhadeep Roy
- Department of Pharmacology and Toxicology, National Institute of Pharmaceutical Education and Research, Kolkata, West Bengal 700054, India.
| | - Manjari Singh
- Department of Pharmaceutical Sciences, Assam Central University, Silchar, Assam 788011, India.
| |
Collapse
|
10
|
Yuan Z, Li Y, Zhang S, Wang X, Dou H, Yu X, Zhang Z, Yang S, Xiao M. Extracellular matrix remodeling in tumor progression and immune escape: from mechanisms to treatments. Mol Cancer 2023; 22:48. [PMID: 36906534 PMCID: PMC10007858 DOI: 10.1186/s12943-023-01744-8] [Citation(s) in RCA: 154] [Impact Index Per Article: 154.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2023] [Accepted: 02/11/2023] [Indexed: 03/13/2023] Open
Abstract
The malignant tumor is a multi-etiological, systemic and complex disease characterized by uncontrolled cell proliferation and distant metastasis. Anticancer treatments including adjuvant therapies and targeted therapies are effective in eliminating cancer cells but in a limited number of patients. Increasing evidence suggests that the extracellular matrix (ECM) plays an important role in tumor development through changes in macromolecule components, degradation enzymes and stiffness. These variations are under the control of cellular components in tumor tissue via the aberrant activation of signaling pathways, the interaction of the ECM components to multiple surface receptors, and mechanical impact. Additionally, the ECM shaped by cancer regulates immune cells which results in an immune suppressive microenvironment and hinders the efficacy of immunotherapies. Thus, the ECM acts as a barrier to protect cancer from treatments and supports tumor progression. Nevertheless, the profound regulatory network of the ECM remodeling hampers the design of individualized antitumor treatment. Here, we elaborate on the composition of the malignant ECM, and discuss the specific mechanisms of the ECM remodeling. Precisely, we highlight the impact of the ECM remodeling on tumor development, including proliferation, anoikis, metastasis, angiogenesis, lymphangiogenesis, and immune escape. Finally, we emphasize ECM "normalization" as a potential strategy for anti-malignant treatment.
Collapse
Affiliation(s)
- Zhennan Yuan
- Department of Oncological Surgery, Harbin Medical University Cancer Hospital, Harbin, 150081, China
| | - Yingpu Li
- Department of Oncological Surgery, Harbin Medical University Cancer Hospital, Harbin, 150081, China
| | - Sifan Zhang
- Department of Neurobiology, Harbin Medical University, Harbin, 150081, China
| | - Xueying Wang
- Department of Otolaryngology Head and Neck Surgery, Xiangya Hospital, Central South University, Changsha, 410008, China
| | - He Dou
- Department of Oncological Surgery, Harbin Medical University Cancer Hospital, Harbin, 150081, China
| | - Xi Yu
- Department of Gynecological Oncology, Harbin Medical University Cancer Hospital, Harbin, 150081, China
| | - Zhiren Zhang
- NHC Key Laboratory of Cell Transplantation, The First Affiliated Hospital of Harbin Medical University, Harbin, 150001, China.,Institute of Metabolic Disease, Heilongjiang Academy of Medical Science, Heilongjiang Key Laboratory for Metabolic Disorder and Cancer Related Cardiovascular Diseases, Harbin, 150001, China
| | - Shanshan Yang
- Department of Gynecological Radiotherapy, Harbin Medical University Cancer Hospital, Harbin, 150000, China.
| | - Min Xiao
- Department of Oncological Surgery, Harbin Medical University Cancer Hospital, Harbin, 150081, China.
| |
Collapse
|
11
|
Karrobi K, Tank A, Fuzail MA, Kalidoss M, Tilbury K, Zaman M, Ferruzzi J, Roblyer D. Fluorescence Lifetime Imaging Microscopy (FLIM) reveals spatial-metabolic changes in 3D breast cancer spheroids. Sci Rep 2023; 13:3624. [PMID: 36869092 PMCID: PMC9984376 DOI: 10.1038/s41598-023-30403-7] [Citation(s) in RCA: 7] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2022] [Accepted: 02/22/2023] [Indexed: 03/05/2023] Open
Abstract
Cancer cells are mechanically sensitive to physical properties of the microenvironment, which can affect downstream signaling to promote malignancy, in part through the modulation of metabolic pathways. Fluorescence Lifetime Imaging Microscopy (FLIM) can be used to measure the fluorescence lifetime of endogenous fluorophores, such as the metabolic co-factors NAD(P)H and FAD, in live samples. We used multiphoton FLIM to investigate the changes in cellular metabolism of 3D breast spheroids derived from MCF-10A and MD-MB-231 cell lines embedded in collagen with varying densities (1 vs. 4 mg/ml) over time (Day 0 vs. Day 3). MCF-10A spheroids demonstrated spatial gradients, with the cells closest to the spheroid edge exhibiting FLIM changes consistent with a shift towards oxidative phosphorylation (OXPHOS) while the spheroid core had changes consistent with a shift towards glycolysis. The MDA-MB-231 spheroids had a large shift consistent with increased OXPHOS with a more pronounced change at the higher collagen concentration. The MDA-MB-231 spheroids invaded into the collagen gel over time and cells that traveled the farthest had the largest changes consistent with a shift towards OXPHOS. Overall, these results suggest that the cells in contact with the extracellular matrix (ECM) and those that migrated the farthest had changes consistent with a metabolic shift towards OXPHOS. More generally, these results demonstrate the ability of multiphoton FLIM to characterize how spheroids metabolism and spatial metabolic gradients are modified by physical properties of the 3D ECM.
Collapse
Affiliation(s)
- Kavon Karrobi
- Department of Biomedical Engineering, Boston University, Boston, MA, 02215, USA
| | - Anup Tank
- Department of Biomedical Engineering, Boston University, Boston, MA, 02215, USA
| | | | - Madhumathi Kalidoss
- Department of Biomedical Engineering, Boston University, Boston, MA, 02215, USA
| | - Karissa Tilbury
- Department of Chemical and Biomedical Engineering, University of Maine, Orono, ME, 04469, USA
| | - Muhammad Zaman
- Department of Biomedical Engineering, Boston University, Boston, MA, 02215, USA
| | - Jacopo Ferruzzi
- Department of Biomedical Engineering, Boston University, Boston, MA, 02215, USA
- Department of Biomedical Engineering, The University of Texas at Dallas, Richardson, TX, 75080, USA
| | - Darren Roblyer
- Department of Biomedical Engineering, Boston University, Boston, MA, 02215, USA.
| |
Collapse
|
12
|
Nguyen HM, Paulishak W, Oladejo M, Wood L. Dynamic tumor microenvironment, molecular heterogeneity, and distinct immunologic portrait of triple-negative breast cancer: an impact on classification and treatment approaches. Breast Cancer 2023; 30:167-186. [PMID: 36399321 DOI: 10.1007/s12282-022-01415-4] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2022] [Accepted: 10/26/2022] [Indexed: 11/19/2022]
Abstract
Heterogeneity of the tumor microenvironment (TME) and the lack of a definite targetable receptor in triple-negative breast cancer (TNBC) has carved a niche for this cancer as a particularly therapeutically challenging form of breast cancer. However, recent advances in high-throughput genomic analysis have provided new insights into the unique microenvironment and defining characteristics of various subsets of TNBC. This improved understanding has contributed to the development of novel therapeutic strategies including targeted therapies such as PARP inhibitors and CDK inhibitors. Moreover, the recent FDA approval of the immune checkpoint inhibitor against programmed cell death protein 1 (PD-1), pembrolizumab and atezolizumab, holds the promise of improving the quality of life and increasing the overall survival of TNBC patients. This recent approval is one of the many therapeutically novel strategies that are currently being exploited in clinical trials toward eventual contribution to the oncologist's toolbox against TNBC. In this review, we comprehensively discuss TNBC's distinct TME and its immunophenotype. Furthermore, we highlight the histological and molecular classification of this cancer. More importantly, we describe how these characteristics and classifications contribute to the current standards of care and how they steer the development of newer and more targeted therapies toward achieving peak therapeutic goals in the treatment of TNBC.
Collapse
Affiliation(s)
- Hong-My Nguyen
- Department of Immunotherapeutics and Biotechnology, Texas Tech University Health Sciences Center, Jerry H. Hodge School of Pharmacy, Abilene, TX, 79601, USA
| | - Wyatt Paulishak
- Department of Immunotherapeutics and Biotechnology, Texas Tech University Health Sciences Center, Jerry H. Hodge School of Pharmacy, Abilene, TX, 79601, USA
| | - Mariam Oladejo
- Department of Immunotherapeutics and Biotechnology, Texas Tech University Health Sciences Center, Jerry H. Hodge School of Pharmacy, Abilene, TX, 79601, USA
| | - Laurence Wood
- Department of Immunotherapeutics and Biotechnology, Texas Tech University Health Sciences Center, Jerry H. Hodge School of Pharmacy, Abilene, TX, 79601, USA.
| |
Collapse
|
13
|
Liu H, Yin H, Wang Z, Yuan Q, Xu F, Chen Y, Li C. Rho A/ROCK1 signaling-mediated metabolic reprogramming of valvular interstitial cells toward Warburg effect accelerates aortic valve calcification via AMPK/RUNX2 axis. Cell Death Dis 2023; 14:108. [PMID: 36774349 PMCID: PMC9922265 DOI: 10.1038/s41419-023-05642-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2022] [Revised: 01/29/2023] [Accepted: 02/01/2023] [Indexed: 02/13/2023]
Abstract
The aberrant differentiation of valvular interstitial cells (VICs) to osteogenic lineages promotes calcified aortic valves disease (CAVD), partly activated by potentially destructive hemodynamic forces. These involve Rho A/ROCK1 signaling, a mechano-sensing pathway. However, how Rho A/ROCK1 signaling transduces mechanical signals into cellular responses and disrupts normal VIC homeostasis remain unclear. We examined Rho A/ROCK1 signaling in human aortic valves, and further detected how Rho A/ROCK1 signaling regulates mineralization in human VICs. Aortic valves (CAVD n = 22, normal control (NC) n = 12) from patients undergoing valve replacement were investigated. Immunostaining and western blotting analysis indicated that Rho A/ROCK1 signaling, as well as key transporters and enzymes involved in the Warburg effect, were markedly upregulated in human calcified aortic valves compared with those in the controls. In vitro, Rho A/ROCK1-induced calcification was confirmed as AMPK-dependent, via a mechanism involving metabolic reprogramming of human VICs to Warburg effect. Y-27632, a selective ROCK1 inhibitor, suppressed the Warburg effect, rescued AMPK activity and subsequently increased RUNX2 ubiquitin-proteasome degradation, leading to decreased RUNX2 protein accumulation in human VICs under pathological osteogenic stimulus. Rho A/ROCK1 signaling, which is elevated in human calcified aortic valves, plays a positive role in valvular calcification, partially through its ability to drive metabolic switching of VICs to the Warburg effect, leading to altered AMPK activity and RUNX2 protein accumulation. Thus, Rho A/ROCK1 signaling could be an important and unrecognized hub of destructive hemodynamics and cellular aerobic glycolysis that is essential to promote the CAVD process.
Collapse
Affiliation(s)
- Huiruo Liu
- Department of Emergency Medicine and Chest Pain Center, Qilu Hospital of Shandong University, Jinan, Shandong, China
- Key Laboratory of Emergency and Critical Care Medicine of Shandong Province, Qilu Hospital of Shandong University, Jinan, Shandong, China
- The Key Laboratory of Cardiovascular Remodeling and Function Research, Chinese Ministry of Education and Chinese Ministry of Public Health, Qilu Hospital of Shandong University, Jinan, Shandong, China
| | - Hang Yin
- Department of Emergency Medicine and Chest Pain Center, Qilu Hospital of Shandong University, Jinan, Shandong, China
- Key Laboratory of Emergency and Critical Care Medicine of Shandong Province, Qilu Hospital of Shandong University, Jinan, Shandong, China
- The Key Laboratory of Cardiovascular Remodeling and Function Research, Chinese Ministry of Education and Chinese Ministry of Public Health, Qilu Hospital of Shandong University, Jinan, Shandong, China
| | - Zhen Wang
- Department of Emergency Medicine and Chest Pain Center, Qilu Hospital of Shandong University, Jinan, Shandong, China
- Key Laboratory of Emergency and Critical Care Medicine of Shandong Province, Qilu Hospital of Shandong University, Jinan, Shandong, China
- The Key Laboratory of Cardiovascular Remodeling and Function Research, Chinese Ministry of Education and Chinese Ministry of Public Health, Qilu Hospital of Shandong University, Jinan, Shandong, China
| | - Qiuhuan Yuan
- Department of Emergency Medicine and Chest Pain Center, Qilu Hospital of Shandong University, Jinan, Shandong, China
- Key Laboratory of Emergency and Critical Care Medicine of Shandong Province, Qilu Hospital of Shandong University, Jinan, Shandong, China
- The Key Laboratory of Cardiovascular Remodeling and Function Research, Chinese Ministry of Education and Chinese Ministry of Public Health, Qilu Hospital of Shandong University, Jinan, Shandong, China
| | - Feng Xu
- Department of Emergency Medicine and Chest Pain Center, Qilu Hospital of Shandong University, Jinan, Shandong, China
- Key Laboratory of Emergency and Critical Care Medicine of Shandong Province, Qilu Hospital of Shandong University, Jinan, Shandong, China
- The Key Laboratory of Cardiovascular Remodeling and Function Research, Chinese Ministry of Education and Chinese Ministry of Public Health, Qilu Hospital of Shandong University, Jinan, Shandong, China
| | - Yuguo Chen
- Department of Emergency Medicine and Chest Pain Center, Qilu Hospital of Shandong University, Jinan, Shandong, China.
- Key Laboratory of Emergency and Critical Care Medicine of Shandong Province, Qilu Hospital of Shandong University, Jinan, Shandong, China.
- The Key Laboratory of Cardiovascular Remodeling and Function Research, Chinese Ministry of Education and Chinese Ministry of Public Health, Qilu Hospital of Shandong University, Jinan, Shandong, China.
| | - Chuanbao Li
- Department of Emergency Medicine and Chest Pain Center, Qilu Hospital of Shandong University, Jinan, Shandong, China.
- Key Laboratory of Emergency and Critical Care Medicine of Shandong Province, Qilu Hospital of Shandong University, Jinan, Shandong, China.
- The Key Laboratory of Cardiovascular Remodeling and Function Research, Chinese Ministry of Education and Chinese Ministry of Public Health, Qilu Hospital of Shandong University, Jinan, Shandong, China.
| |
Collapse
|
14
|
Lien CH, Chen ZH, Phan QH. Birefringence effect studies of collagen formed by nonenzymatic glycation using dual-retarder Mueller polarimetry. JOURNAL OF BIOMEDICAL OPTICS 2022; 27:087001. [PMID: 36452033 PMCID: PMC9349470 DOI: 10.1117/1.jbo.27.8.087001] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 03/16/2022] [Accepted: 07/18/2022] [Indexed: 06/17/2023]
Abstract
SIGNIFICANCE Nonenzymatic glycation of collagen covalently attaches an addition of sugar molecules that initially were involved in a reversibly reaction with amino groups on the protein. Due to the ultimate formation of stable irreversible advanced glycation end products, the process of glycation leads to abnormal irreversible cross-linking, which ultimately accumulates with age and/or diabetes in the extracellular matrix, altering its organization. AIM We report the use of dual-retarder Mueller polarimetry in conjunction with phase retardance to differentiate collagen cross-linking in a normal collagen gel matrix from that in tissues with nonenzymatic cross-linking. APPROACH A dual-liquid crystal-based Mueller polarimetry system that involves electronic modulation of polarization state generators (PSGs) was employed to produce all types of polarization states without moving any part and enable detection of the signal directly using a Stokes polarimeter. The linear phase retardance response was obtained for the characterization of the solution and gel forms of collagen using differential Mueller matrix analysis. RESULTS We found that linear phase retardance measurements via differential Mueller matrix polarimetry successfully differentiated collagen gel matrices with different degrees of cross-linking formed by a nonenzymatic glycation process and demonstrated that this technology constitutes a quick and simple modality. CONCLUSIONS This approach has high sensitivity for studying differences in fibrillar cross-linking in glycated collagen. Further, our work suggests that this method of structural analysis has potential clinical diagnostic value owing to its noninvasive and cost-efficient nature.
Collapse
Affiliation(s)
- Chi-Hsiang Lien
- National United University, Department of Mechanical Engineering, Miaoli, Taiwan
| | - Zong-Hong Chen
- National United University, Department of Mechanical Engineering, Miaoli, Taiwan
| | - Quoc-Hung Phan
- National United University, Department of Mechanical Engineering, Miaoli, Taiwan
| |
Collapse
|
15
|
A bioinformatic analysis of WFDC2 (HE4) expression in high grade serous ovarian cancer reveals tumor-specific changes in metabolic and extracellular matrix gene expression. Med Oncol 2022; 39:71. [PMID: 35568777 PMCID: PMC9107348 DOI: 10.1007/s12032-022-01665-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2021] [Accepted: 01/22/2022] [Indexed: 10/31/2022]
Abstract
Human epididymis protein-4 (HE4/WFDC2) has been well-studied as an ovarian cancer clinical biomarker. To improve our understanding of its functional role in high grade serous ovarian cancer, we determined transcriptomic differences between ovarian tumors with high- versus low-WFDC2 mRNA levels in The Cancer Genome Atlas dataset. High-WFDC2 transcript levels were significantly associated with reduced survival in stage III/IV serous ovarian cancer patients. Differential expression and correlation analyses revealed secretory leukocyte peptidase inhibitor (SLPI/WFDC4) as the gene most positively correlated with WFDC2, while A kinase anchor protein-12 was most negatively correlated. WFDC2 and SLPI were strongly correlated across many cancers. Gene ontology analysis revealed enrichment of oxidative phosphorylation in differentially expressed genes associated with high-WFDC2 levels, while extracellular matrix organization was enriched among genes associated with low-WFDC2 levels. Immune cell subsets found to be positively correlated with WFDC2 levels were B cells and plasmacytoid dendritic cells, while neutrophils and endothelial cells were negatively correlated with WFDC2. Results were compared with DepMap cell culture gene expression data. Gene ontology analysis of k-means clustering revealed that genes associated with low-WFDC2 were also enriched in extracellular matrix and adhesion categories, while high-WFDC2 genes were enriched in epithelial cell proliferation and peptidase activity. These results support previous findings regarding the effect of HE4/WFDC2 on ovarian cancer pathogenesis in cell lines and mouse models, while adding another layer of complexity to its potential functions in ovarian tumor tissue. Further experimental explorations of these findings in the context of the tumor microenvironment are merited.
Collapse
|
16
|
Peltanová B, Holcová Polanská H, Raudenská M, Balvan J, Navrátil J, Vičar T, Gumulec J, Čechová B, Kräter M, Guck J, Kalfeřt D, Grega M, Plzák J, Betka J, Masařík M. mRNA Subtype of Cancer-Associated Fibroblasts Significantly Affects Key Characteristics of Head and Neck Cancer Cells. Cancers (Basel) 2022; 14:2286. [PMID: 35565415 PMCID: PMC9102192 DOI: 10.3390/cancers14092286] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2022] [Revised: 04/27/2022] [Accepted: 04/27/2022] [Indexed: 12/10/2022] Open
Abstract
Head and neck squamous cell carcinomas (HNSCC) belong among severe and highly complex malignant diseases showing a high level of heterogeneity and consequently also a variance in therapeutic response, regardless of clinical stage. Our study implies that the progression of HNSCC may be supported by cancer-associated fibroblasts (CAFs) in the tumour microenvironment (TME) and the heterogeneity of this disease may lie in the level of cooperation between CAFs and epithelial cancer cells, as communication between CAFs and epithelial cancer cells seems to be a key factor for the sustained growth of the tumour mass. In this study, we investigated how CAFs derived from tumours of different mRNA subtypes influence the proliferation of cancer cells and their metabolic and biomechanical reprogramming. We also investigated the clinicopathological significance of the expression of these metabolism-related genes in tissue samples of HNSCC patients to identify a possible gene signature typical for HNSCC progression. We found that the right kind of cooperation between cancer cells and CAFs is needed for tumour growth and progression, and only specific mRNA subtypes can support the growth of primary cancer cells or metastases. Specifically, during coculture, cancer cell colony supporting effect and effect of CAFs on cell stiffness of cancer cells are driven by the mRNA subtype of the tumour from which the CAFs are derived. The degree of colony-forming support is reflected in cancer cell glycolysis levels and lactate shuttle-related transporters.
Collapse
Affiliation(s)
- Barbora Peltanová
- Department of Pathological Physiology, Faculty of Medicine, Masaryk University, Kamenice 5, 62500 Brno, Czech Republic; (B.P.); (H.H.P.); (M.R.); (J.B.); (J.N.); (J.G.); (B.Č.)
- Department of Physiology, Faculty of Medicine, Masaryk University, Kamenice 5, 62500 Brno, Czech Republic;
| | - Hana Holcová Polanská
- Department of Pathological Physiology, Faculty of Medicine, Masaryk University, Kamenice 5, 62500 Brno, Czech Republic; (B.P.); (H.H.P.); (M.R.); (J.B.); (J.N.); (J.G.); (B.Č.)
- Department of Physiology, Faculty of Medicine, Masaryk University, Kamenice 5, 62500 Brno, Czech Republic;
| | - Martina Raudenská
- Department of Pathological Physiology, Faculty of Medicine, Masaryk University, Kamenice 5, 62500 Brno, Czech Republic; (B.P.); (H.H.P.); (M.R.); (J.B.); (J.N.); (J.G.); (B.Č.)
- Department of Physiology, Faculty of Medicine, Masaryk University, Kamenice 5, 62500 Brno, Czech Republic;
- Department of Chemistry and Biochemistry, Mendel University in Brno, Zemedelska 1, 61300 Brno, Czech Republic
| | - Jan Balvan
- Department of Pathological Physiology, Faculty of Medicine, Masaryk University, Kamenice 5, 62500 Brno, Czech Republic; (B.P.); (H.H.P.); (M.R.); (J.B.); (J.N.); (J.G.); (B.Č.)
- Department of Physiology, Faculty of Medicine, Masaryk University, Kamenice 5, 62500 Brno, Czech Republic;
| | - Jiří Navrátil
- Department of Pathological Physiology, Faculty of Medicine, Masaryk University, Kamenice 5, 62500 Brno, Czech Republic; (B.P.); (H.H.P.); (M.R.); (J.B.); (J.N.); (J.G.); (B.Č.)
| | - Tomáš Vičar
- Department of Physiology, Faculty of Medicine, Masaryk University, Kamenice 5, 62500 Brno, Czech Republic;
| | - Jaromír Gumulec
- Department of Pathological Physiology, Faculty of Medicine, Masaryk University, Kamenice 5, 62500 Brno, Czech Republic; (B.P.); (H.H.P.); (M.R.); (J.B.); (J.N.); (J.G.); (B.Č.)
| | - Barbora Čechová
- Department of Pathological Physiology, Faculty of Medicine, Masaryk University, Kamenice 5, 62500 Brno, Czech Republic; (B.P.); (H.H.P.); (M.R.); (J.B.); (J.N.); (J.G.); (B.Č.)
| | - Martin Kräter
- Max Planck Institute for the Science of Light, Staudtstraße 2, 91058 Erlangen, Germany; (M.K.); (J.G.)
| | - Jochen Guck
- Max Planck Institute for the Science of Light, Staudtstraße 2, 91058 Erlangen, Germany; (M.K.); (J.G.)
| | - David Kalfeřt
- Department of Otorhinolaryngology and Head and Neck Surgery, First Faculty of Medicine, University Hospital Motol, Charles University, V Uvalu 84, 15006 Prague, Czech Republic; (D.K.); (J.P.); (J.B.)
| | - Marek Grega
- Department of Pathology and Molecular Medicine, 2nd Faculty of Medicine, University Hospital Motol, Charles University, V Uvalu 84, 15006 Prague, Czech Republic;
| | - Jan Plzák
- Department of Otorhinolaryngology and Head and Neck Surgery, First Faculty of Medicine, University Hospital Motol, Charles University, V Uvalu 84, 15006 Prague, Czech Republic; (D.K.); (J.P.); (J.B.)
| | - Jan Betka
- Department of Otorhinolaryngology and Head and Neck Surgery, First Faculty of Medicine, University Hospital Motol, Charles University, V Uvalu 84, 15006 Prague, Czech Republic; (D.K.); (J.P.); (J.B.)
| | - Michal Masařík
- Department of Pathological Physiology, Faculty of Medicine, Masaryk University, Kamenice 5, 62500 Brno, Czech Republic; (B.P.); (H.H.P.); (M.R.); (J.B.); (J.N.); (J.G.); (B.Č.)
- Department of Physiology, Faculty of Medicine, Masaryk University, Kamenice 5, 62500 Brno, Czech Republic;
- BIOCEV, First Faculty of Medicine, Charles University, Prumyslova 595, 25250 Vestec, Czech Republic
| |
Collapse
|
17
|
Fabiano E, Zhang J, Reinhart-King C. Tissue density in the progression of breast cancer: Bedside to bench and back again. CURRENT OPINION IN BIOMEDICAL ENGINEERING 2022; 22. [DOI: 10.1016/j.cobme.2022.100383] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
|
18
|
Cancer-associated fibroblasts promote oral squamous cell carcinoma progression through LOX-mediated matrix stiffness. J Transl Med 2021; 19:513. [PMID: 34930321 PMCID: PMC8686394 DOI: 10.1186/s12967-021-03181-x] [Citation(s) in RCA: 28] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2021] [Accepted: 12/03/2021] [Indexed: 02/08/2023] Open
Abstract
Background Cancer-associated fibroblasts (CAFs), the most abundant cells in the tumor microenvironment, have prominent roles in the development of solid tumors as stromal targets. However, the underlying mechanism of CAFs’ function in oral squamous cell carcinoma (OSCC) development remains unclear. Here, we investigated the role of lysyl oxidase (LOX) expression in CAFs in tumor stromal remodeling and the mechanism of its effect on OSCC progression. Methods Multiple immunohistochemistry (IHC) staining was performed to detect the correlation of CAFs and LOX in the stroma of OSCC specimens, as well as the correlation with clinicopathological parameters and prognosis. The expression of LOX in CAFs were detected by RT-qPCR and western blot. The effects of LOX in CAFs on the biological characteristics of OSCC cell line were investigated using CCK-8, wound-healing and transwell assay. CAFs were co-cultured with type I collagen in vitro, and collagen contraction test, microstructure observation and rheometer were used to detect the effect of CAFs on remodeling collagen matrix. Then, collagen with different stiffness were established to investigate the effect of matrix stiffness on the progression of OSCC. Moreover, we used focal adhesion kinase (FAK) phosphorylation inhibitors to explored whether the increase in matrix stiffness promote the progression of OSCC through activating FAK phosphorylation pathway. Results LOX was colocalized with CAFs in the stroma of OSCC tissues, and its expression was significantly related to the degree of malignant differentiation and poor prognosis in OSCC. LOX was highly expressed in CAFs, and its knockdown impaired the proliferation, migration, invasion and EMT process of OSCC cells. The expression of LOX in CAFs can catalyze collagen crosslinking and increase matrix stiffness. Furthermore, CAFs-derived LOX-mediated increase in collagen stiffness induced morphological changes and promoted invasion and EMT process in OSCC cells by activating FAK phosphorylation pathway. Conclusions Our findings suggest that CAFs highly express LOX in the stroma of OSCC and can remodel the matrix collagen microenvironment, and the increase in matrix stiffness mediated by CAFs-derived LOX promotes OSCC development through FAK phosphorylation pathway. Thus, LOX may be a potential target for the early diagnosis and therapeutic treatment of OSCC. Supplementary Information The online version contains supplementary material available at 10.1186/s12967-021-03181-x.
Collapse
|
19
|
Sarwar M, Sykes PH, Chitcholtan K, Evans JJ. Collagen I dysregulation is pivotal for ovarian cancer progression. Tissue Cell 2021; 74:101704. [PMID: 34871826 DOI: 10.1016/j.tice.2021.101704] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2021] [Revised: 11/23/2021] [Accepted: 11/24/2021] [Indexed: 11/16/2022]
Abstract
As a principal matrisomal protein, collagen is involved in the regulation of the structural framework of extracellular matrix (ECM) and therefore is potentially crucial in determining the biophysical character of the ECM. It has been suggested that collagen architecture plays a role in ovarian cancer development, progression and therapeutic responses which led us to examine the collagen morphology in normal and cancerous ovarian tissue. Also, the behaviour of ovarian cancer cells cultured in four qualitatively different collagen gels was investigated. The results here provide evidence that collagen I morphology in the cancerous ovary is distinct from that in the normal ovary. Tumour-associated collagen I showed streams or channels of thick elongated collagen I fibrils. Moreover, fibril alignment was significantly more prevalent in endometrioid and clear cell cancers than other ovarian cancer subtypes. In this work, for the first-time collagen I architecture profiling (CAP) was introduced using histochemical staining, which distinguished between the collagen I morphologies of ovarian cancer subtypes. Immunohistochemical examination of ovarian normal and cancerous tissues also supported the notion that focal adhesion and Rho signalling are upregulated in ovarian cancers, especially in the high-grade serous tumours, as indicated by higher expression of p-FAK and p190RhoGEF. The results also support the concept that collagen I architecture, which might be collagen I concentration-dependent, influences proliferation in ovarian cancer cells. The study provides evidence that modification of collagen I architecture integrity is associated with ovarian cancer development and therapeutic responses.
Collapse
Affiliation(s)
- Makhdoom Sarwar
- Department of Obstetrics and Gynaecology, University of Otago, Christchurch, 2 Riccarton Avenue, Christchurch, 8011, New Zealand.
| | - Peter H Sykes
- Department of Obstetrics and Gynaecology, University of Otago, Christchurch, 2 Riccarton Avenue, Christchurch, 8011, New Zealand
| | - Kenny Chitcholtan
- Department of Obstetrics and Gynaecology, University of Otago, Christchurch, 2 Riccarton Avenue, Christchurch, 8011, New Zealand
| | - John J Evans
- Department of Obstetrics and Gynaecology, University of Otago, Christchurch, 2 Riccarton Avenue, Christchurch, 8011, New Zealand; MacDiarmid Institute of Advanced Materials and Nanotechnology, Christchurch, New Zealand
| |
Collapse
|
20
|
Kang W, Ferruzzi J, Spatarelu CP, Han YL, Sharma Y, Koehler SA, Mitchel JA, Khan A, Butler JP, Roblyer D, Zaman MH, Park JA, Guo M, Chen Z, Pegoraro AF, Fredberg JJ. A novel jamming phase diagram links tumor invasion to non-equilibrium phase separation. iScience 2021; 24:103252. [PMID: 34755092 PMCID: PMC8564056 DOI: 10.1016/j.isci.2021.103252] [Citation(s) in RCA: 37] [Impact Index Per Article: 12.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2021] [Revised: 09/14/2021] [Accepted: 10/07/2021] [Indexed: 12/31/2022] Open
Abstract
It is well established that the early malignant tumor invades surrounding extracellular matrix (ECM) in a manner that depends upon material properties of constituent cells, surrounding ECM, and their interactions. Recent studies have established the capacity of the invading tumor spheroids to evolve into coexistent solid-like, fluid-like, and gas-like phases. Using breast cancer cell lines invading into engineered ECM, here we show that the spheroid interior develops spatial and temporal heterogeneities in material phase which, depending upon cell type and matrix density, ultimately result in a variety of phase separation patterns at the invasive front. Using a computational approach, we further show that these patterns are captured by a novel jamming phase diagram. We suggest that non-equilibrium phase separation based upon jamming and unjamming transitions may provide a unifying physical picture to describe cellular migratory dynamics within, and invasion from, a tumor.
Collapse
Affiliation(s)
- Wenying Kang
- Department of Environmental Science, Harvard T.H. Chan School of Public Health, Boston, MA 02115, USA
| | - Jacopo Ferruzzi
- Department of Biomedical Engineering, Boston University, Boston, MA 02215, USA
- Department of Bioengineering, University of Texas at Dallas, Richardson, TX 75080, USA
| | | | - Yu Long Han
- Department of Mechanical Engineering, Massachusetts Institute of Technology, Cambridge, MA 02139, USA
| | - Yasha Sharma
- Department of Environmental Science, Harvard T.H. Chan School of Public Health, Boston, MA 02115, USA
| | - Stephan A. Koehler
- Department of Environmental Science, Harvard T.H. Chan School of Public Health, Boston, MA 02115, USA
| | - Jennifer A. Mitchel
- Department of Environmental Science, Harvard T.H. Chan School of Public Health, Boston, MA 02115, USA
| | - Adil Khan
- Department of Bioengineering, University of Texas at Dallas, Richardson, TX 75080, USA
| | - James P. Butler
- Department of Environmental Science, Harvard T.H. Chan School of Public Health, Boston, MA 02115, USA
- Department of Medicine, Brigham and Women’s Hospital and Harvard Medical School, Boston, MA 02115, USA
| | - Darren Roblyer
- Department of Biomedical Engineering, Boston University, Boston, MA 02215, USA
| | - Muhammad H. Zaman
- Department of Biomedical Engineering, Boston University, Boston, MA 02215, USA
- Howard Hughes Medical Institute, Boston University, Boston, MA 02115, USA
| | - Jin-Ah Park
- Department of Environmental Science, Harvard T.H. Chan School of Public Health, Boston, MA 02115, USA
| | - Ming Guo
- Department of Mechanical Engineering, Massachusetts Institute of Technology, Cambridge, MA 02139, USA
| | - Zi Chen
- Thayer School of Engineering, Dartmouth College, Hanover, NH 03755, USA
| | | | - Jeffrey J. Fredberg
- Department of Environmental Science, Harvard T.H. Chan School of Public Health, Boston, MA 02115, USA
| |
Collapse
|
21
|
Sthijns MMJPE, van Blitterswijk CA, LaPointe VLS. Synthetic Materials that Affect the Extracellular Matrix via Cellular Metabolism and Responses to a Metabolic State. Front Bioeng Biotechnol 2021; 9:742132. [PMID: 34708025 PMCID: PMC8542861 DOI: 10.3389/fbioe.2021.742132] [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: 07/15/2021] [Accepted: 09/28/2021] [Indexed: 11/18/2022] Open
Abstract
In regenerative medicine and tissue engineering, many materials are developed to mimic the extracellular matrix (ECM). However, these ECM-mimicking materials do not yet completely recapitulate the diversity and complexity of biological tissue-specific ECM. In this review, an alternative strategy is proposed to generate ECM, namely synthesizing a material that functions as a drug delivery system, releasing molecules that target cellular metabolic pathways and thereby stimulate the local cells to create their own ECM. This is based on the fact that ECM synthesis, modification, composition, signaling, stiffness, and degradation are modulated by cellular metabolism. Metabolism can be targeted at different levels, ranging from modulating the availability of substrates or co-factors to regulating the activity of essential transcription factors. Depending on the drug of interest, its characteristics, mechanism of action, cellular target, and application, a different drug delivery system should be designed. Metabolic drugs modulating the ECM require cellular uptake for their function, therefore reversible linkers are recommended. Preferably the metabolic modulators are only released when needed, which will be upon a specific metabolic state, a change in ECM stiffness, or ECM remodeling. Therefore, reversible linkers that respond to an environmental stimulus could be incorporated. All in all, a novel strategy is suggested to develop a tissue-specific ECM by generating a synthetic material that releases metabolic molecules modulating the ECM. Various ways to modulate the ECM properties via the metabolism are reviewed and guidelines for the development of these materials are provided.
Collapse
Affiliation(s)
- Mireille M J P E Sthijns
- Department of Cell Biology-Inspired Tissue Engineering, MERLN Institute for Technology-Inspired Regenerative Medicine, Maastricht University, Maastricht, Netherlands.,Department of Food Innovation and Health at the Centre of Healthy Eating and Food Innovation, Maastricht University, Maastricht, Netherlands
| | - Clemens A van Blitterswijk
- Department of Cell Biology-Inspired Tissue Engineering, MERLN Institute for Technology-Inspired Regenerative Medicine, Maastricht University, Maastricht, Netherlands
| | - Vanessa L S LaPointe
- Department of Cell Biology-Inspired Tissue Engineering, MERLN Institute for Technology-Inspired Regenerative Medicine, Maastricht University, Maastricht, Netherlands
| |
Collapse
|
22
|
Bisht VS, Giri K, Kumar D, Ambatipudi K. Oxygen and metabolic reprogramming in the tumor microenvironment influences metastasis homing. Cancer Biol Ther 2021; 22:493-512. [PMID: 34696706 DOI: 10.1080/15384047.2021.1992233] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/20/2022] Open
Abstract
Tumor metastasis is the leading cause of cancer mortality, often characterized by abnormal cell growth and invasion to distant organs. The cancer invasion due to epithelial to mesenchymal transition is affected by metabolic and oxygen availability in the tumor-associated micro-environment. A precise alteration in oxygen and metabolic signaling between healthy and metastatic cells is a substantial probe for understanding tumor progression and metastasis. Molecular heterogeneity in the tumor microenvironment help to sustain the metastatic cell growth during their survival shift from low to high metabolic-oxygen-rich sites and reinforces the metastatic events. This review highlighted the crucial role of oxygen and metabolites in metastatic progression and exemplified the role of metabolic rewiring and oxygen availability in cancer cell adaptation. Furthermore, we have also addressed potential applications of altered oxygen and metabolic networking with tumor type that could be a signature pattern to assess tumor growth and chemotherapeutics efficacy in managing cancer metastasis.
Collapse
Affiliation(s)
- Vinod S Bisht
- Department of Biosciences and Bioengineering, Indian Institute of Technology Roorkee, Roorkee, India
| | - Kuldeep Giri
- Department of Biosciences and Bioengineering, Indian Institute of Technology Roorkee, Roorkee, India
| | - Deepak Kumar
- Department of Cancer Biology, Central Drug Research Institute, Lucknow, India.,Academy of Scientific & Innovative Research, New Delhi, India
| | - Kiran Ambatipudi
- Department of Biosciences and Bioengineering, Indian Institute of Technology Roorkee, Roorkee, India
| |
Collapse
|
23
|
Mosier JA, Schwager SC, Boyajian DA, Reinhart-King CA. Cancer cell metabolic plasticity in migration and metastasis. Clin Exp Metastasis 2021; 38:343-359. [PMID: 34076787 DOI: 10.1007/s10585-021-10102-1] [Citation(s) in RCA: 40] [Impact Index Per Article: 13.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2020] [Accepted: 05/08/2021] [Indexed: 12/13/2022]
Abstract
Metabolic reprogramming is a hallmark of cancer metastasis in which cancer cells manipulate their metabolic profile to meet the dynamic energetic requirements of the tumor microenvironment. Though cancer cell proliferation and migration through the extracellular matrix are key steps of cancer progression, they are not necessarily fueled by the same metabolites and energy production pathways. The two main metabolic pathways cancer cells use to derive energy from glucose, glycolysis and oxidative phosphorylation, are preferentially and plastically utilized by cancer cells depending on both their intrinsic metabolic properties and their surrounding environment. Mechanical factors in the microenvironment, such as collagen density, pore size, and alignment, and biochemical factors, such as oxygen and glucose availability, have been shown to influence both cell migration and glucose metabolism. As cancer cells have been identified as preferentially utilizing glycolysis or oxidative phosphorylation based on heterogeneous intrinsic or extrinsic factors, the relationship between cancer cell metabolism and metastatic potential is of recent interest. Here, we review current in vitro and in vivo findings in the context of cancer cell metabolism during migration and metastasis and extrapolate potential clinical applications of this work that could aid in diagnosing and tracking cancer progression in vivo by monitoring metabolism. We also review current progress in the development of a variety of metabolically targeted anti-metastatic drugs, both in clinical trials and approved for distribution, and highlight potential routes for incorporating our recent understanding of metabolic plasticity into therapeutic directions. By further understanding cancer cell energy production pathways and metabolic plasticity, more effective and successful clinical imaging and therapeutics can be developed to diagnose, target, and inhibit metastasis.
Collapse
Affiliation(s)
- Jenna A Mosier
- Department of Biomedical Engineering, Vanderbilt University, Nashville, TN, USA
| | - Samantha C Schwager
- Department of Biomedical Engineering, Vanderbilt University, Nashville, TN, USA
| | - David A Boyajian
- Department of Radiology, Weill Cornell Medical College, New York, NY, USA
| | | |
Collapse
|
24
|
Zanotelli MR, Zhang J, Reinhart-King CA. Mechanoresponsive metabolism in cancer cell migration and metastasis. Cell Metab 2021; 33:1307-1321. [PMID: 33915111 PMCID: PMC9015673 DOI: 10.1016/j.cmet.2021.04.002] [Citation(s) in RCA: 144] [Impact Index Per Article: 48.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/25/2021] [Revised: 03/16/2021] [Accepted: 04/05/2021] [Indexed: 12/12/2022]
Abstract
Altered tissue mechanics and metabolism are defining characteristics of cancer that impact not only proliferation but also migration. While migrating through a mechanically and spatially heterogeneous microenvironment, changes in metabolism allow cells to dynamically tune energy generation and bioenergetics in response to fluctuating energy needs. Physical cues from the extracellular matrix influence mechanosignaling pathways, cell mechanics, and cytoskeletal architecture to alter presentation and function of metabolic enzymes. In cancer, altered mechanosensing and metabolic reprogramming supports metabolic plasticity and high energy production while cells migrate and metastasize. Here, we discuss the role of mechanoresponsive metabolism in regulating cell migration and supporting metastasis as well as the potential of therapeutically targeting cancer metabolism to block motility and potentially metastasis.
Collapse
Affiliation(s)
- Matthew R Zanotelli
- Nancy E. and Peter C. Meinig School of Biomedical Engineering, Cornell University, Ithaca, NY 14853, USA; Department of Biomedical Engineering, Vanderbilt University, Nashville, TN 37235, USA
| | - Jian Zhang
- Department of Biomedical Engineering, Vanderbilt University, Nashville, TN 37235, USA
| | | |
Collapse
|
25
|
Cong ATQ, Pimenta RML, Holy J, Heikal AA. Associated anisotropy of intrinsic NAD(P)H for monitoring changes in the metabolic activities of breast cancer cells (4T1) in three-dimensional collagen matrix. Phys Chem Chem Phys 2021; 23:12692-12705. [PMID: 34036961 DOI: 10.1039/d0cp06635d] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
Abstract
The majority of in vitro studies of living cells are routinely conducted in a two-dimensional (2D) monolayer culture. Recent studies, however, suggest that 2D cell culture promotes specific types of aberrant cell behaviors due to the growth on non-physiologically stiff surfaces and the lack of the tissue-based extracellular matrix. Here, we investigate the sensitivity of the two-photon (2P) rotational dynamics of the intrinsic reduced nicotinamide adenine dinucleotide (phosphate), NAD(P)H, to changes in the metabolic state of the metastatic murine breast cancer cells (4T1) in 2D monolayer and three-dimensional (3D) collagen matrix cultures. Time-resolved 2P-associated anisotropy measurements reveal that the rotational dynamics of free and enzyme-bound NAD(P)H in 4T1 cells are correlated to changes in the metabolic state of 2D and 3D cell cultures. In addition to the type of cell culture, we also investigated the metabolic response of 4T1 cells to treatment with two metabolic inhibitors (MD1 and TPPBr). The statistical analyses of our results enabled us to identify which of the fitting parameters of the observed time-resolved associate anisotropy of cellular NAD(P)H were significantly sensitive to changes in the metabolic state of 4T1 cells. Using a black-box model, the population fractions of free and bound NAD(P)H were used to estimate the corresponding equilibrium constant and the standard Gibbs free energy changes that are associated with underlying metabolic pathways of 4T1 cells in 2D and 3D cultures. These rotational dynamics analyses are in agreement with the standard 2P-fluorescence lifetime imaging microscopy (FLIM) measurements on the same cell line, cell cultures, and metabolic inhibition. These studies represent an important step towards the development of a noninvasive, time-resolved associated anisotropy to complement 2P-FLIM in order to elucidate the underlying cellular metabolism and metabolic plasticity in more complex in vivo, tumor-like models using intrinsic NADH autofluorescence.
Collapse
Affiliation(s)
- Anh T Q Cong
- Department of Chemistry and Biochemistry, Swenson College of Science and Engineering, University of Minnesota Duluth, 1039 University Drive, Duluth, MN 55812, USA.
| | | | | | | |
Collapse
|
26
|
Wu Y, Zanotelli MR, Zhang J, Reinhart-King CA. Matrix-driven changes in metabolism support cytoskeletal activity to promote cell migration. Biophys J 2021; 120:1705-1717. [PMID: 33705759 PMCID: PMC8204337 DOI: 10.1016/j.bpj.2021.02.044] [Citation(s) in RCA: 19] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2020] [Revised: 02/03/2021] [Accepted: 02/23/2021] [Indexed: 01/21/2023] Open
Abstract
The microenvironment provides both active and passive mechanical cues that regulate cell morphology, adhesion, migration, and metabolism. Although the cellular response to those mechanical cues often requires energy-intensive actin cytoskeletal remodeling and actomyosin contractility, it remains unclear how cells dynamically adapt their metabolic activity to altered mechanical cues to support migration. Here, we investigated the changes in cellular metabolic activity in response to different two-dimensional and three-dimensional microenvironmental conditions and how these changes relate to cytoskeletal activity and migration. Utilizing collagen micropatterning on polyacrylamide gels, intracellular energy levels and oxidative phosphorylation were found to be correlated with cell elongation and spreading and necessary for membrane ruffling. To determine whether this relationship holds in more physiological three-dimensional matrices, collagen matrices were used to show that intracellular energy state was also correlated with protrusive activity and increased with matrix density. Pharmacological inhibition of oxidative phosphorylation revealed that cancer cells rely on oxidative phosphorylation to meet the elevated energy requirements for protrusive activity and migration in denser matrices. Together, these findings suggest that mechanical regulation of cytoskeletal activity during spreading and migration by the physical microenvironment is driven by an altered metabolic profile.
Collapse
Affiliation(s)
- Yusheng Wu
- Department of Biomedical Engineering, Vanderbilt University, Nashville, Tennessee
| | - Matthew R Zanotelli
- Department of Biomedical Engineering, Vanderbilt University, Nashville, Tennessee; Nancy E. and Peter C. Meinig School of Biomedical Engineering, Cornell University, Ithaca, New York
| | - Jian Zhang
- Department of Biomedical Engineering, Vanderbilt University, Nashville, Tennessee
| | | |
Collapse
|
27
|
Haensel D, Jin S, Sun P, Cinco R, Dragan M, Nguyen Q, Cang Z, Gong Y, Vu R, MacLean AL, Kessenbrock K, Gratton E, Nie Q, Dai X. Defining Epidermal Basal Cell States during Skin Homeostasis and Wound Healing Using Single-Cell Transcriptomics. Cell Rep 2021; 30:3932-3947.e6. [PMID: 32187560 PMCID: PMC7218802 DOI: 10.1016/j.celrep.2020.02.091] [Citation(s) in RCA: 119] [Impact Index Per Article: 39.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2019] [Revised: 01/07/2020] [Accepted: 02/25/2020] [Indexed: 01/17/2023] Open
Abstract
Our knowledge of transcriptional heterogeneities in epithelial stem and progenitor cell compartments is limited. Epidermal basal cells sustain cutaneous tissue maintenance and drive wound healing. Previous studies have probed basal cell heterogeneity in stem and progenitor potential, but a comprehensive dissection of basal cell dynamics during differentiation is lacking. Using single-cell RNA sequencing coupled with RNAScope and fluorescence lifetime imaging, we identify three non-proliferative and one proliferative basal cell state in homeostatic skin that differ in metabolic preference and become spatially partitioned during wound re-epithelialization. Pseudotemporal trajectory and RNA velocity analyses predict a quasi-linear differentiation hierarchy where basal cells progress from Col17a1Hi/Trp63Hi state to early-response state, proliferate at the juncture of these two states, or become growth arrested before differentiating into spinous cells. Wound healing induces plasticity manifested by dynamic basal-spinous interconversions at multiple basal transcriptional states. Our study provides a systematic view of epidermal cellular dynamics, supporting a revised “hierarchical-lineage” model of homeostasis. Haensel et al. performed a comprehensive dissection of the cellular makeup of skin during homeostasis and wound healing and the molecular heterogeneity and cellular dynamics within its stem-cell-containing epidermal basal layer. Their work provides insights and stimulates further investigation into the mechanism of skin maintenance and repair.
Collapse
Affiliation(s)
- Daniel Haensel
- Department of Biological Chemistry, School of Medicine, University of California, Irvine, CA 92697, USA
- The NSF-Simons Center for Multiscale Cell Fate Research, University of California, Irvine, CA 92627, USA
- These authors contributed equally
| | - Suoqin Jin
- Department of Mathematics, University of California, Irvine, CA 92697, USA
- These authors contributed equally
| | - Peng Sun
- Department of Biological Chemistry, School of Medicine, University of California, Irvine, CA 92697, USA
| | - Rachel Cinco
- Laboratory for Fluorescence Dynamics, Department of Biomedical Engineering, University of California, Irvine, CA 92697, USA
| | - Morgan Dragan
- Department of Biological Chemistry, School of Medicine, University of California, Irvine, CA 92697, USA
- The NSF-Simons Center for Multiscale Cell Fate Research, University of California, Irvine, CA 92627, USA
| | - Quy Nguyen
- Department of Biological Chemistry, School of Medicine, University of California, Irvine, CA 92697, USA
| | - Zixuan Cang
- The NSF-Simons Center for Multiscale Cell Fate Research, University of California, Irvine, CA 92627, USA
- Department of Mathematics, University of California, Irvine, CA 92697, USA
| | - Yanwen Gong
- Department of Biological Chemistry, School of Medicine, University of California, Irvine, CA 92697, USA
- Center for Complex Biological Systems, University of California, Irvine, CA 92697, USA
| | - Remy Vu
- Department of Biological Chemistry, School of Medicine, University of California, Irvine, CA 92697, USA
- The NSF-Simons Center for Multiscale Cell Fate Research, University of California, Irvine, CA 92627, USA
| | - Adam L. MacLean
- Department of Mathematics, University of California, Irvine, CA 92697, USA
| | - Kai Kessenbrock
- Department of Biological Chemistry, School of Medicine, University of California, Irvine, CA 92697, USA
| | - Enrico Gratton
- Laboratory for Fluorescence Dynamics, Department of Biomedical Engineering, University of California, Irvine, CA 92697, USA
| | - Qing Nie
- The NSF-Simons Center for Multiscale Cell Fate Research, University of California, Irvine, CA 92627, USA
- Department of Mathematics, University of California, Irvine, CA 92697, USA
- Department of Developmental and Cell Biology, University of California, Irvine, CA 92697, USA
- Correspondence: (Q.N.), (X.D.)
| | - Xing Dai
- Department of Biological Chemistry, School of Medicine, University of California, Irvine, CA 92697, USA
- The NSF-Simons Center for Multiscale Cell Fate Research, University of California, Irvine, CA 92627, USA
- Lead Contact
- Correspondence: (Q.N.), (X.D.)
| |
Collapse
|
28
|
DeWane G, Salvi AM, DeMali KA. Fueling the cytoskeleton - links between cell metabolism and actin remodeling. J Cell Sci 2021; 134:jcs248385. [PMID: 33558441 PMCID: PMC7888749 DOI: 10.1242/jcs.248385] [Citation(s) in RCA: 61] [Impact Index Per Article: 20.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022] Open
Abstract
Attention has long focused on the actin cytoskeleton as a unit capable of organizing into ensembles that control cell shape, polarity, migration and the establishment of intercellular contacts that support tissue architecture. However, these investigations do not consider observations made over 40 years ago that the actin cytoskeleton directly binds metabolic enzymes, or emerging evidence suggesting that the rearrangement and assembly of the actin cytoskeleton is a major energetic drain. This Review examines recent studies probing how cells adjust their metabolism to provide the energy necessary for cytoskeletal remodeling that occurs during cell migration, epithelial to mesenchymal transitions, and the cellular response to external forces. These studies have revealed that mechanotransduction, cell migration, and epithelial to mesenchymal transitions are accompanied by alterations in glycolysis and oxidative phosphorylation. These metabolic changes provide energy to support the actin cytoskeletal rearrangements necessary to allow cells to assemble the branched actin networks required for cell movement and epithelial to mesenchymal transitions and the large actin bundles necessary for cells to withstand forces. In this Review, we discuss the emerging evidence suggesting that the regulation of these events is highly complex with metabolism affecting the actin cytoskeleton and vice versa.
Collapse
Affiliation(s)
- Gillian DeWane
- Department of Biochemistry, University of Iowa Carver College of Medicine, Iowa City, IA 52246, USA
| | - Alicia M Salvi
- Department of Biochemistry, University of Iowa Carver College of Medicine, Iowa City, IA 52246, USA
| | - Kris A DeMali
- Department of Biochemistry, University of Iowa Carver College of Medicine, Iowa City, IA 52246, USA
| |
Collapse
|
29
|
Karta J, Bossicard Y, Kotzamanis K, Dolznig H, Letellier E. Mapping the Metabolic Networks of Tumor Cells and Cancer-Associated Fibroblasts. Cells 2021; 10:304. [PMID: 33540679 PMCID: PMC7912987 DOI: 10.3390/cells10020304] [Citation(s) in RCA: 22] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2020] [Revised: 01/20/2021] [Accepted: 01/26/2021] [Indexed: 12/12/2022] Open
Abstract
Metabolism is considered to be the core of all cellular activity. Thus, extensive studies of metabolic processes are ongoing in various fields of biology, including cancer research. Cancer cells are known to adapt their metabolism to sustain high proliferation rates and survive in unfavorable environments with low oxygen and nutrient concentrations. Hence, targeting cancer cell metabolism is a promising therapeutic strategy in cancer research. However, cancers consist not only of genetically altered tumor cells but are interwoven with endothelial cells, immune cells and fibroblasts, which together with the extracellular matrix (ECM) constitute the tumor microenvironment (TME). Cancer-associated fibroblasts (CAFs), which are linked to poor prognosis in different cancer types, are one important component of the TME. CAFs play a significant role in reprogramming the metabolic landscape of tumor cells, but how, and in what manner, this interaction takes place remains rather unclear. This review aims to highlight the metabolic landscape of tumor cells and CAFs, including their recently identified subtypes, in different tumor types. In addition, we discuss various in vitro and in vivo metabolic techniques as well as different in silico computational tools that can be used to identify and characterize CAF-tumor cell interactions. Finally, we provide our view on how mapping the complex metabolic networks of stromal-tumor metabolism will help in finding novel metabolic targets for cancer treatment.
Collapse
Affiliation(s)
- Jessica Karta
- Molecular Disease Mechanisms Group, Department of Life Sciences and Medicine, Faculty of Science, Technology and Medicine, University of Luxembourg, 6 avenue du Swing, L-4367 Belval, Luxembourg; (J.K.); (Y.B.); (K.K.)
| | - Ysaline Bossicard
- Molecular Disease Mechanisms Group, Department of Life Sciences and Medicine, Faculty of Science, Technology and Medicine, University of Luxembourg, 6 avenue du Swing, L-4367 Belval, Luxembourg; (J.K.); (Y.B.); (K.K.)
| | - Konstantinos Kotzamanis
- Molecular Disease Mechanisms Group, Department of Life Sciences and Medicine, Faculty of Science, Technology and Medicine, University of Luxembourg, 6 avenue du Swing, L-4367 Belval, Luxembourg; (J.K.); (Y.B.); (K.K.)
| | - Helmut Dolznig
- Tumor Stroma Interaction Group, Institute of Medical Genetics, Medical University of Vienna, Währinger Strasse 10, 1090 Vienna, Austria;
| | - Elisabeth Letellier
- Molecular Disease Mechanisms Group, Department of Life Sciences and Medicine, Faculty of Science, Technology and Medicine, University of Luxembourg, 6 avenue du Swing, L-4367 Belval, Luxembourg; (J.K.); (Y.B.); (K.K.)
| |
Collapse
|
30
|
Liao Y, You R, Fan M, Feng S, Lu D, Lu Y. Determination of NADH by Surface Enhanced Raman Scattering Using Au@MB@Ag NPs. Aust J Chem 2021. [DOI: 10.1071/ch21178] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/23/2022]
Abstract
Nicotinamide adenine dinucleotide (NADH) is an important coenzyme involved in various metabolic processes of living cells. As an important biomarker, NADH is associated with breast cancer and Alzheimer’s disease. In this paper, silver plated gold core–shell nanoparticles containing Raman signal molecules were synthesised on the basis of bare gold. Using the Raman peak corresponding to the 4-mercaptobenzonitrile (MB) silent region C≡N vibration for quantification, while avoiding competition with the precious metal surface binding site to be measured, it can also be free from the interference of endogenous biomolecules. On the one hand, it can correct the working curve, on the other hand, it can avoid competing with the binding site. Compared with the core–shell structure prepared here, the limit of detection (LOD) for NADH was only 10−5 M for bare gold and the LOD for the core–shell structure prepared on the basis of bare gold was 3.3 × 10−7 M. In terms of correction, with Rhodamine 6G (R6G) as a Raman signalling molecule, the R2 value before SERS detection and correction is only 0.9405, and the R2 value after correction increases to 0.9853. The unique fingerprint peak of SERS was used to realise the quantitative detection of NADH, which realizes the detection of NADH in complex biological samples of serum and provides the possibility for expanding the early diagnosis of breast cancer.
Collapse
|
31
|
Jagiełło A, Lim M, Botvinick E. Dermal fibroblasts and triple-negative mammary epithelial cancer cells differentially stiffen their local matrix. APL Bioeng 2020; 4:046105. [PMID: 33305163 PMCID: PMC7719046 DOI: 10.1063/5.0021030] [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: 07/06/2020] [Accepted: 11/18/2020] [Indexed: 02/07/2023] Open
Abstract
The bulk measurement of extracellular matrix (ECM) stiffness is commonly used in mechanobiology. However, past studies by our group show that peri-cellular stiffness is quite heterogeneous and divergent from the bulk. We use optical tweezers active microrheology (AMR) to quantify how two phenotypically distinct migratory cell lines establish dissimilar patterns of peri-cellular stiffness. Dermal fibroblasts (DFs) and triple-negative human breast cancer cells MDA-MB-231 (MDAs) were embedded within type 1 collagen (T1C) hydrogels polymerized at two concentrations: 1.0 mg/ml and 1.5 mg/ml. We found DFs increase the local stiffness of 1.0 mg/ml T1C hydrogels but, surprisingly, do not alter the stiffness of 1.5 mg/ml T1C hydrogels. In contrast, MDAs predominantly do not stiffen T1C hydrogels as compared to cell-free controls. The results suggest that MDAs adapt to the bulk ECM stiffness, while DFs regulate local stiffness to levels they intrinsically prefer. In other experiments, cells were treated with transforming growth factor-β1 (TGF-β1), glucose, or ROCK inhibitor Y27632, which have known effects on DFs and MDAs related to migration, proliferation, and contractility. The results show that TGF-β1 alters stiffness anisotropy, while glucose increases stiffness magnitude around DFs but not MDAs and Y27632 treatment inhibits cell-mediated stiffening. Both cell lines exhibit an elongated morphology and local stiffness anisotropy, where the stiffer axis depends on the cell line, T1C concentration, and treatment. In summary, our findings demonstrate that AMR reveals otherwise masked mechanical properties such as spatial gradients and anisotropy, which are known to affect cell behavior at the macro-scale. The same properties manifest with similar magnitude around single cells.
Collapse
Affiliation(s)
- Alicja Jagiełło
- Department of Biomedical Engineering, University of California Irvine, Irvine, California 92697, USA
| | - Micah Lim
- Department of Biomedical Engineering, University of California Irvine, Irvine, California 92697, USA
| | | |
Collapse
|
32
|
Energy expenditure during cell spreading influences the cellular response to matrix stiffness. Biomaterials 2020; 267:120494. [PMID: 33130323 DOI: 10.1016/j.biomaterials.2020.120494] [Citation(s) in RCA: 35] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2020] [Revised: 10/20/2020] [Accepted: 10/23/2020] [Indexed: 02/07/2023]
Abstract
Cells respond to the mechanical properties of the extracellular matrix (ECM) through formation of focal adhesions (FAs), re-organization of the actin cytoskeleton and adjustment of cell contractility. These are energy-demanding processes, but a potential causality between mechanical cues (matrix stiffness) and cellular (energy) metabolism remains largely unexplored. Here, we cultured human mesenchymal stem cells (hMSCs) on stiff (20 kPa) or soft (1 kPa) substrate and demonstrate that cytoskeletal reorganization and FA formation spreading on stiff substrates lead to a drop in intracellular ATP levels, correlating with activation of AMP-activated protein kinase (AMPK). The resulting increase in ATP levels further facilitates cell spreading and reinforces cell tension of the steady state, and coincides with nuclear localization of YAP/TAZ and Runx2. While on soft substrates (1 kPa), lowered ATP levels limit these cellular mechanoresponses. Furthermore, genetic ablation of AMPK lowered cellular ATP levels on stiff substrate and strongly reduced responses to substrate stiffness. Together, these findings reveal a hitherto unidentified relationship between energy expenditure and the cellular mechanoresponse, and point to AMPK as a key mediator of stem cell fate in response to ECM mechanics.
Collapse
|
33
|
Microenvironment remodeled by tumor and stromal cells elevates fibroblast-derived COL1A1 and facilitates ovarian cancer metastasis. Exp Cell Res 2020; 394:112153. [PMID: 32589888 DOI: 10.1016/j.yexcr.2020.112153] [Citation(s) in RCA: 43] [Impact Index Per Article: 10.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2020] [Revised: 06/08/2020] [Accepted: 06/21/2020] [Indexed: 01/25/2023]
Abstract
Wide peritoneal metastasis is the cause of the highest lethality of ovarian cancer in gynecologic malignancies. Ascites play a key role in ovarian cancer metastasis, but involved mechanism is uncertain. Here, we performed a quantitative proteomics of ascites, and found that collagen type I alpha 1 (COL1A1) was notably elevated in ascites from epithelial ovarian cancer patients compared to normal peritoneal fluids, and verified that elevated COL1A1 was mainly originated from fibroblasts. COL1A1 promoted migration and invasion of ovarian cancer cells, but such effects were partially eliminated by COL1A1 antibodies. Intraperitoneally injected COL1A1 accelerated intraperitoneal metastasis of ovarian cancer xenograft in NOD/SCID mice. Further, COL1A1 activated downstream AKT phosphorylation by binding to membrane surface receptor integrin β1 (ITGB1). Knockdown or blockage of ITGB1 reversed COL1A1 enhanced migration and invasion in ovarian cancer cells. Conversely, ovarian cancer ascites and fibrinogen promoted fibroblasts to secrete COL1A1. Elevated fibrinogen in ascites might be associated with increased vascular permeability induced by ovarian cancer. Our findings suggest that microenvironment remodeled by tumor cells and stromal cells promotes fibroblasts to secrete COL1A1 and facilitates the metastasis of ovarian cancer, which may provide a new approach for ovarian cancer therapeutics.
Collapse
|
34
|
Fong EJ, Strelez C, Mumenthaler SM. A Perspective on Expanding Our Understanding of Cancer Treatments by Integrating Approaches from the Biological and Physical Sciences. SLAS DISCOVERY 2020; 25:672-683. [PMID: 32297829 PMCID: PMC7372587 DOI: 10.1177/2472555220915830] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
Multicellular systems such as cancer suffer from immense complexity. It is imperative to capture the heterogeneity of these systems across scales to achieve a deeper understanding of the underlying biology and develop effective treatment strategies. In this perspective article, we will discuss how recent technologies and approaches from the biological and physical sciences have transformed traditional ways of measuring, interpreting, and treating cancer. During the SLAS 2019 Annual Meeting, SBI2 hosted a Special Interest Group (SIG) on this topic. Academic and industry leaders engaged in discussions surrounding what biological model systems are appropriate to study cancer complexity, what assays are necessary to interrogate this complexity, and how physical sciences approaches may be useful to detangle this complexity. In particular, we examined the utility of mathematical models in predicting cancer progression and treatment response when tightly integrated with reproducible, quantitative, and dynamic biological measurements achieved using high-content imaging and analysis. The dialogue centered around the impetus for convergent biosciences, bringing new perspectives to cancer research to further understand this complex adaptive system and successfully intervene therapeutically.
Collapse
Affiliation(s)
- Emma J Fong
- Lawrence J. Ellison Institute for Transformative Medicine, University of Southern California, Los Angeles, CA, USA
| | - Carly Strelez
- Lawrence J. Ellison Institute for Transformative Medicine, University of Southern California, Los Angeles, CA, USA
| | - Shannon M Mumenthaler
- Lawrence J. Ellison Institute for Transformative Medicine, University of Southern California, Los Angeles, CA, USA
| |
Collapse
|
35
|
Transcriptional diversity and bioenergetic shift in human breast cancer metastasis revealed by single-cell RNA sequencing. Nat Cell Biol 2020; 22:310-320. [PMID: 32144411 DOI: 10.1038/s41556-020-0477-0] [Citation(s) in RCA: 183] [Impact Index Per Article: 45.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2018] [Accepted: 02/04/2020] [Indexed: 12/24/2022]
Abstract
Although metastasis remains the cause of most cancer-related mortality, mechanisms governing seeding in distal tissues are poorly understood. Here, we establish a robust method for the identification of global transcriptomic changes in rare metastatic cells during seeding using single-cell RNA sequencing and patient-derived-xenograft models of breast cancer. We find that both primary tumours and micrometastases display transcriptional heterogeneity but micrometastases harbour a distinct transcriptome program conserved across patient-derived-xenograft models that is highly predictive of poor survival of patients. Pathway analysis revealed mitochondrial oxidative phosphorylation as the top pathway upregulated in micrometastases, in contrast to higher levels of glycolytic enzymes in primary tumour cells, which we corroborated by flow cytometric and metabolomic analyses. Pharmacological inhibition of oxidative phosphorylation dramatically attenuated metastatic seeding in the lungs, which demonstrates the functional importance of oxidative phosphorylation in metastasis and highlights its potential as a therapeutic target to prevent metastatic spread in patients with breast cancer.
Collapse
|
36
|
Hanumantharao SN, Que CA, Vogl BJ, Rao S. Engineered Three-Dimensional Scaffolds Modulating Fate of Breast Cancer Cells Using Stiffness and Morphology Related Cell Adhesion. IEEE OPEN JOURNAL OF ENGINEERING IN MEDICINE AND BIOLOGY 2020; 1:41-48. [PMID: 35402960 PMCID: PMC8979620 DOI: 10.1109/ojemb.2020.2965084] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2019] [Revised: 12/22/2019] [Accepted: 12/23/2019] [Indexed: 11/25/2022] Open
Abstract
Goal: Artificially engineering the tumor microenvironment in vitro as a vital tool for understanding the mechanism of tumor progression. In this study, we developed three-dimensional cell scaffold systems with different topographical features and mechanical properties but similar surface chemistry. The cell behavior was modulated by the topography and mechanical properties of the scaffold. Methods: Adenocarcinoma (MCF7), triple-negative (MDA-MB-231) and premalignant (MCF10AneoT) breast cancer cells were seeded on the scaffold systems. The cell viability, cell-cell interaction and cell-matrix interactions were analyzed. The preferential growth and alignment of specific population of cells were demonstrated. Results: Among the different scaffolds, triple-negative breast cancer cells preferred honeycomb scaffolds while adenocarcinoma cells favored mesh scaffolds and premalignant cells preferred the aligned scaffolds. Conclusions: The 3D model system developed here can be used to support growth of only specific cell populations or for the growth of tumors. This model can be used for understanding the topographical and mechanical features affecting tumorigenesis, cancer cell growth and migration behavior of malignant and metastatic cancer cells.
Collapse
Affiliation(s)
| | | | | | - Smitha Rao
- Department of Biomedical EngineeringMichigan Technological UniversityHoughtonMI49931USA
| |
Collapse
|
37
|
Baltes F, Pfeifer V, Silbermann K, Caspers J, Wantoch von Rekowski K, Schlesinger M, Bendas G. β 1-Integrin binding to collagen type 1 transmits breast cancer cells into chemoresistance by activating ABC efflux transporters. BIOCHIMICA ET BIOPHYSICA ACTA-MOLECULAR CELL RESEARCH 2020; 1867:118663. [PMID: 31987794 DOI: 10.1016/j.bbamcr.2020.118663] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/18/2019] [Revised: 01/20/2020] [Accepted: 01/23/2020] [Indexed: 12/13/2022]
Abstract
Molecular interactions of tumor cells with the microenvironment are regarded as onset of chemotherapy resistance, referred to as cell adhesion mediated drug resistance (CAM-DR). Here we elucidate a mechanism of CAM-DR in breast cancer cells in vitro. We show that human MCF-7 and MDA-MB-231 breast cancer cells decrease their sensitivity towards cisplatin, doxorubicin, and mitoxantrone cytotoxicity upon binding to collagen type 1 (COL1) or fibronectin (FN). The intracellular concentrations of doxorubicin and mitoxantrone were decreased upon cell cultivation on COL1, while cellular cisplatin levels remained unaffected. Since doxorubicin and mitoxantrone are transporter substrates, this refers to ATP binding cassette (ABC) efflux transporter activities. The activation of the transporters BCRP, P-gp and MRP1 was shown by fluorescence assays to distinguish the individual input of these transporters to resistance in presence of COL1 and related to their expression levels by western blot. An ABC transporter inhibitor was able to re-sensitize COL1-treated cells for doxorubicin and mitoxantrone toxicity. Antibody-blocking of β1-integrin (ITGB1) induced sensitization towards the indicated cytostatic drugs by attenuating the increased ABC efflux activity. This refers to a key role of ITGB1 for matrix binding and subsequent transporter activation. A downregulation of α2β1 integrin following COL1 binding appears as clear indication for the relationship between ITGB1 and ABC transporters in regulating resistance formation, while knockdown of ITGB1 leads to a significant upregulation of all three transporters. Our data provide evidence for a role of CAM-DR in breast cancer via an ITGB1 - transporter axis and offer promising therapeutic targets for cancer sensitization.
Collapse
Affiliation(s)
| | | | | | | | | | | | - Gerd Bendas
- Department of Pharmacy, University of Bonn, Germany.
| |
Collapse
|
38
|
Mishriki S, Aithal S, Gupta T, Sahu RP, Geng F, Puri IK. Fibroblasts Accelerate Formation and Improve Reproducibility of 3D Cellular Structures Printed with Magnetic Assistance. RESEARCH (WASHINGTON, D.C.) 2020; 2020:3970530. [PMID: 32776011 PMCID: PMC7395227 DOI: 10.34133/2020/3970530] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 01/30/2020] [Accepted: 06/18/2020] [Indexed: 12/05/2022]
Abstract
Fibroblasts (mouse, NIH/3T3) are combined with MDA-MB-231 cells to accelerate the formation and improve the reproducibility of 3D cellular structures printed with magnetic assistance. Fibroblasts and MDA-MB-231 cells are cocultured to produce 12.5 : 87.5, 25 : 75, and 50 : 50 total population mixtures. These mixtures are suspended in a cell medium containing a paramagnetic salt, Gd-DTPA, which increases the magnetic susceptibility of the medium with respect to the cells. A 3D monotypic MDA-MB-231 cellular structure is printed within 24 hours with magnetic assistance, whereas it takes 48 hours to form a similar structure through gravitational settling alone. The maximum projected areas and circularities, and cellular ATP levels of the printed structures are measured for 336 hours. Increasing the relative amounts of the fibroblasts mixed with the MDA-MB-231 cells decreases the time taken to form the structures and improves their reproducibility. Structures produced through gravitational settling have larger maximum projected areas and cellular ATP, but are deemed less reproducible. The distribution of individual cell lines in the cocultured 3D cellular structures shows that printing with magnetic assistance yields 3D cellular structures that resemble in vivo tumors more closely than those formed through gravitational settling. The results validate our hypothesis that (1) fibroblasts act as a "glue" that supports the formation of 3D cellular structures, and (2) the structures are produced more rapidly and with greater reproducibility with magnetically assisted printing than through gravitational settling alone. Printing of 3D cellular structures with magnetic assistance has applications relevant to drug discovery, lab-on-chip devices, and tissue engineering.
Collapse
Affiliation(s)
- Sarah Mishriki
- School of Biomedical Engineering, McMaster University, Hamilton, Ontario, Canada
| | - Srivatsa Aithal
- Department of Mechanical Engineering, McMaster University, Hamilton, Ontario, Canada
| | - Tamaghna Gupta
- School of Biomedical Engineering, McMaster University, Hamilton, Ontario, Canada
| | - Rakesh P. Sahu
- Department of Mechanical Engineering, McMaster University, Hamilton, Ontario, Canada
- Department of Materials Science and Engineering, McMaster University, Hamilton, Ontario, Canada
| | - Fei Geng
- Department of Mechanical Engineering, McMaster University, Hamilton, Ontario, Canada
- Walter Booth School of Engineering Practice and Technology, Hamilton, Ontario, Canada
| | - Ishwar K. Puri
- School of Biomedical Engineering, McMaster University, Hamilton, Ontario, Canada
- Department of Mechanical Engineering, McMaster University, Hamilton, Ontario, Canada
- Department of Materials Science and Engineering, McMaster University, Hamilton, Ontario, Canada
| |
Collapse
|
39
|
Li J, Wang X, Zheng K, Liu Y, Li J, Wang S, Liu K, Song X, Li N, Xie S, Wang S. The clinical significance of collagen family gene expression in esophageal squamous cell carcinoma. PeerJ 2019; 7:e7705. [PMID: 31598423 PMCID: PMC6779144 DOI: 10.7717/peerj.7705] [Citation(s) in RCA: 21] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2019] [Accepted: 08/19/2019] [Indexed: 12/24/2022] Open
Abstract
BACKGROUND Esophageal squamous cell carcinoma (ESCC) is a subtype of esophageal cancer with high incidence and mortality. Due to the poor 5-year survival rates of patients with ESCC, exploring novel diagnostic markers for early ESCC is emergent. Collagen, the abundant constituent of extracellular matrix, plays a critical role in tumor growth and epithelial-mesenchymal transition. However, the clinical significance of collagen genes in ESCC has been rarely studied. In this work, we systematically analyzed the gene expression of whole collagen family in ESCC, aiming to search for ideal biomarkers. METHODS Clinical data and gene expression profiles of ESCC patients were collected from The Cancer Genome Atlas and the gene expression omnibus databases. Bioinformatics methods, including differential expression analysis, survival analysis, gene sets enrichment analysis (GSEA) and co-expression network analysis, were performed to investigate the correlation between the expression patterns of 44 collagen family genes and the development of ESCC. RESULTS A total of 22 genes of collagen family were identified as differentially expressed genes in both the two datasets. Among them, COL1A1, COL10A1 and COL11A1 were particularly up-regulated in ESCC tissues compared to normal controls, while COL4A4, COL6A5 and COL14A1 were notably down-regulated. Besides, patients with low COL6A5 expression or high COL18A1 expression showed poor survival. In addition, a 7-gene prediction model was established based on collagen gene expression to predict patient survival, which had better predictive accuracy than the tumor-node-metastasis staging based model. Finally, GSEA results suggested that collagen genes might be tightly associated with PI3K/Akt/mTOR pathway, p53 pathway, apoptosis, cell cycle, etc. CONCLUSION Several collagen genes could be potential diagnostic and prognostic biomarkers for ESCC. Moreover, a novel 7-gene prediction model is probably useful for predicting survival outcomes of ESCC patients. These findings may facilitate early detection of ESCC and help improves prognosis of the patients.
Collapse
Affiliation(s)
- Jieling Li
- School of Pharmaceutical Sciences, Shenzhen University Health Science Center, Shenzhen, China
| | - Xiao Wang
- Department of Pharmacy, The Second Clinical Medical College (Shenzhen People’s Hospital), Jinan University, Shenzhen, China
| | - Kai Zheng
- School of Pharmaceutical Sciences, Shenzhen University Health Science Center, Shenzhen, China
| | - Ying Liu
- Department of Pharmacy, The Second Clinical Medical College (Shenzhen People’s Hospital), Jinan University, Shenzhen, China
| | - Junjun Li
- School of Pharmaceutical Sciences, Shenzhen University Health Science Center, Shenzhen, China
| | - Shaoqi Wang
- Department of Oncology, Hubei Provincial Corps Hospital, Chinese People Armed Police Forces, Wuhan, China
| | - Kaisheng Liu
- Department of Pharmacy, The Second Clinical Medical College (Shenzhen People’s Hospital), Jinan University, Shenzhen, China
| | - Xun Song
- School of Pharmaceutical Sciences, Shenzhen University Health Science Center, Shenzhen, China
| | - Nan Li
- Department of Pharmacy, The Second Clinical Medical College (Shenzhen People’s Hospital), Jinan University, Shenzhen, China
| | - Shouxia Xie
- Department of Pharmacy, The Second Clinical Medical College (Shenzhen People’s Hospital), Jinan University, Shenzhen, China
| | - Shaoxiang Wang
- School of Pharmaceutical Sciences, Shenzhen University Health Science Center, Shenzhen, China
| |
Collapse
|
40
|
Ma C, Kuzma ML, Bai X, Yang J. Biomaterial-Based Metabolic Regulation in Regenerative Engineering. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2019; 6:1900819. [PMID: 31592416 PMCID: PMC6774061 DOI: 10.1002/advs.201900819] [Citation(s) in RCA: 37] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/09/2019] [Revised: 05/26/2019] [Indexed: 05/22/2023]
Abstract
Recent advances in cell metabolism studies have deepened the appreciation of the role of metabolic regulation in influencing cell behavior during differentiation, angiogenesis, and immune response in the regenerative engineering scenarios. However, the understanding of whether the intracellular metabolic pathways could be influenced by material-derived cues remains limited, although it is now well appreciated that material cues modulate cell functions. Here, an overview of how the regulation of different aspect of cell metabolism, including energy homeostasis, oxygen homeostasis, and redox homeostasis could contribute to modulation of cell function is provided. Furthermore, recent evidence demonstrating how material cues, including the release of inherent metabolic factors (e.g., ions, regulatory metabolites, and oxygen), tuning of the biochemical cues (e.g., inherent antioxidant properties, cell adhesivity, and chemical composition of nanomaterials), and changing in biophysical cues (topography and surface stiffness), may impact cell metabolism toward modulated cell behavior are discussed. Based on the resurgence of interest in cell metabolism and metabolic regulation, further development of biomaterials enabling metabolic regulation toward dictating cell function is poised to have substantial implications for regenerative engineering.
Collapse
Affiliation(s)
- Chuying Ma
- Department of Biomedical EngineeringMaterials Research InstituteThe Huck Institutes of the Life SciencesThe Pennsylvania State UniversityUniversity ParkPA16802USA
| | - Michelle L. Kuzma
- Department of Biomedical EngineeringMaterials Research InstituteThe Huck Institutes of the Life SciencesThe Pennsylvania State UniversityUniversity ParkPA16802USA
| | - Xiaochun Bai
- Academy of OrthopedicsGuangdong ProvinceProvincial Key Laboratory of Bone and Joint Degenerative DiseasesThe Third Affiliated Hospital of Southern Medical UniversityGuangzhou510280China
- Department of Cell BiologyKey Laboratory of Mental Health of the Ministry of EducationSchool of Basic Medical SciencesSouthern Medical UniversityGuangzhou510515China
| | - Jian Yang
- Department of Biomedical EngineeringMaterials Research InstituteThe Huck Institutes of the Life SciencesThe Pennsylvania State UniversityUniversity ParkPA16802USA
| |
Collapse
|
41
|
Abstract
Myosin 2 plays a central role in numerous, fundamental, actin-based biological processes, including cell migration, cell division, and the adhesion of cells to substrates and other cells. Here, we highlight recent studies in which the forces created by actomyosin 2 have been shown to also impact tension-sensitive ion channels and cell metabolism.
Collapse
Affiliation(s)
- Melissa A Quintanilla
- Cell and Molecular Physiology, Loyola University Chicago, Stritch School of Medicine, Center for Translational Research and Education, Maywood, IL, USA
| | - John A Hammer
- Cell and Developmental Biology Center, National Heart Lung and Blood Institute, National Institutes of Health, Bethesda, MD, USA
| | - Jordan R Beach
- Cell and Molecular Physiology, Loyola University Chicago, Stritch School of Medicine, Center for Translational Research and Education, Maywood, IL, USA
| |
Collapse
|
42
|
The Role of Sodium Hydrogen Exchanger 1 in Dysregulation of Proton Dynamics and Reprogramming of Cancer Metabolism as a Sequela. Int J Mol Sci 2019; 20:ijms20153694. [PMID: 31357694 PMCID: PMC6696090 DOI: 10.3390/ijms20153694] [Citation(s) in RCA: 21] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2019] [Revised: 07/23/2019] [Accepted: 07/25/2019] [Indexed: 12/15/2022] Open
Abstract
Cancer cells have an unusual regulation of hydrogen ion dynamics that are driven by poor vascularity perfusion, regional hypoxia, and increased glycolysis. All these forces synergize/orchestrate together to create extracellular acidity and intracellular alkalinity. Precisely, they lead to extracellular pH (pHe) values as low as 6.2 and intracellular pH values as high as 8. This unique pH gradient (∆pHi to ∆pHe) across the cell membrane increases as the tumor progresses, and is markedly displaced from the electrochemical equilibrium of protons. These unusual pH dynamics influence cancer cell biology, including proliferation, metastasis, and metabolic adaptation. Warburg metabolism with increased glycolysis, even in the presence of Oxygen with the subsequent reduction in Krebs’ cycle, is a common feature of most cancers. This metabolic reprogramming confers evolutionary advantages to cancer cells by enhancing their resistance to hypoxia, to chemotherapy or radiotherapy, allowing rapid production of biological building blocks that support cellular proliferation, and shielding against damaging mitochondrial free radicals. In this article, we highlight the interconnected roles of dysregulated pH dynamics in cancer initiation, progression, adaptation, and in determining the programming and re-programming of tumor cell metabolism.
Collapse
|
43
|
Anderson G. Breast cancer: Occluded role of mitochondria N-acetylserotonin/melatonin ratio in co-ordinating pathophysiology. Biochem Pharmacol 2019; 168:259-268. [PMID: 31310736 DOI: 10.1016/j.bcp.2019.07.014] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2019] [Accepted: 07/10/2019] [Indexed: 12/11/2022]
Abstract
A plethora of factors contribute to the biochemical underpinnings of breast cancer, in the absence of any clear, integrative framework. This article proposes that melatonergic pathway regulation within mitochondria provides an integrative framework for the wide array of data driving breast cancer pathophysiology. As melatonin is toxic to breast cancer cells, its production within mitochondria poses a significant challenge to breast cancer cell survival. Consequently, the diverse plasticity in breast cancer cells may arise from a requirement to decrease mitochondria melatonin synthesis. The aryl hydrocarbon receptor role in breast cancer pathophysiology may be mediated by an increase in cytochrome P450 (CYP)1b1 in mitochondria, leading to the backward conversion of melatonin to N-acetylserotonin (NAS). NAS has distinct effects to melatonin, including its activation of the tyrosine receptor kinase B (TrkB) receptor. TrkB activation significantly contributes to breast cancer cell survival and migration. However, the most important aspect of NAS induction by CYP1b1 in breast cancer cells is the prevention of melatonin effects in mitochondria. Many of the changes occurring in breast cancer cells arise from the need to regulate this pathway in mitochondria, allowing this to provide a framework that integrates a host of previously disparate data, including: microRNAs, estrogen, 14-3-3 proteins, sirtuins, glycolysis, oxidative phosphorylation, indoleamine 2,3-dioxygenase and the kynurenine pathways. It is also proposed that this framework provides a pathoetiological model incorporating the early developmental regulation of the gut microbiome that integrates breast cancer risk factors, including obesity. This has significant treatment, prevention and research implications.
Collapse
Affiliation(s)
- George Anderson
- CRC Scotland & London, Eccleston Square, London SW1V 1PH, UK.
| |
Collapse
|
44
|
Chacko JV, Eliceiri KW. Autofluorescence lifetime imaging of cellular metabolism: Sensitivity toward cell density, pH, intracellular, and intercellular heterogeneity. Cytometry A 2019; 95:56-69. [PMID: 30296355 PMCID: PMC6329636 DOI: 10.1002/cyto.a.23603] [Citation(s) in RCA: 37] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2018] [Revised: 08/14/2018] [Accepted: 08/20/2018] [Indexed: 12/14/2022]
Abstract
Autofluorescence imaging (AFI) has greatly accelerated in the last decade, way past its origins in detecting endogenous signals in biological tissues to identify differences between samples. There are many endogenous fluorescence sources of contrast but the most robust and widely utilized have been those associated with metabolism. The intrinsically fluorescent metabolic cofactors nicotinamide adenine dinucleotide (NAD+ /NADH) and flavin adenine dinucleotide (FAD/FADH2 ) have been utilized in a number of AFI applications including basic research, clinical, and pharmaceutical studies. Fluorescence lifetime imaging microscopy (FLIM) has emerged as one of the more powerful AFI tools for NADH and FAD characterization due to its unique ability to noninvasively detect metabolite bound and free states and quantitate cellular redox ratio. However, despite this widespread biological use, many standardization methods are still needed to extend FLIM-based AFI into a fully robust research and clinical diagnostic tools. FLIM is sensitive to a wide range of factors in the fluorophore microenvironment, and there are a number of analysis variables as well. To this end, there has been an emphasis on developing imaging standards and ways to make the image acquisition and analysis more consistent. However, biological conditions during FLIM-based AFI imaging are rarely considered as key sources of FLIM variability. Here, we present several experimental factors with supporting data of the cellular microenvironment such as confluency, pH, inter-/intracellular heterogeneity, and choice of cell line that need to be considered for accurate quantitative FLIM-based AFI measurement of cellular metabolism. © 2018 International Society for Advancement of Cytometry.
Collapse
Affiliation(s)
- Jenu V. Chacko
- Laboratory for Optical and Computational Instrumentation, University of Wisconsin at Madison, Madison WI, USA
| | - Kevin W. Eliceiri
- Laboratory for Optical and Computational Instrumentation, University of Wisconsin at Madison, Madison WI, USA
- Biomedical Engineering Department, University of Wisconsin at Madison, Madison WI, USA
- Morgridge Institute for Research, Madison WI, USA
| |
Collapse
|