1
|
Malakpour-Permlid A, Rodriguez MM, Untracht GR, Andersen PE, Oredsson S, Boisen A, Zór K. High-throughput non-homogenous 3D polycaprolactone scaffold for cancer cell and cancer-associated fibroblast mini-tumors to evaluate drug treatment response. Toxicol Rep 2025; 14:101863. [PMID: 39758801 PMCID: PMC11699757 DOI: 10.1016/j.toxrep.2024.101863] [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: 10/01/2024] [Revised: 12/03/2024] [Accepted: 12/11/2024] [Indexed: 01/07/2025] Open
Abstract
High-throughput screening (HTS) three-dimensional (3D) tumor models are a promising approach for cancer drug discovery, as they more accurately replicate in vivo cell behavior than two-dimensional (2D) models. However, assessing and comparing current 3D models for drug efficacy remains essential, given the significant influence of cellular conditions on treatment response. To develop in vivo mimicking 3D models, we evaluated two HTS 3D models established in 96-well plates with 3D polycaprolactone (PCL) scaffolds fabricated using two distinct methods, resulting in scaffolds with either homogenous or non-homogenous fiber networks. These models, based on human HeLa cervical cancer cells and cancer-associated fibroblasts (CAFs) cultured as mono- or co-cultures within the 3D scaffolds, revealed that anticancer drug paclitaxel (PTX) exhibited consistently higher inhibitory concentration 50 (IC50) in 3D (≥ 1000 nM) compared to 2D (≥ 100 nM), indicating reduced toxicity on cells cultured in 3D. Interestingly, the toxicity of PTX was significantly lower on mini-tumors in non-homogenous 3D (IC50: 600 or 1000 nM) than in homogenous 3D cultures (IC50 exceeding 1000 nM). Microscopic studies revealed that the non-homogenous scaffolds closely resemble the tumor collagen network than their homogeneous counterpart. Both 3D scaffolds offer optimal pore size, facilitating efficient cell infiltration into the depth of 58.1 ± 1.2 µm (homogenous) and 86.4 ± 9.8 µm (non-homogenous) within 3D cultures. Cells cultured in the 3D non-homogenous systems exhibited drug treatment responses closer to in vivo conditions, highlighting the role of scaffold structure and design on cellular response to drug treatment. The PCL-based 3D models provide a robust, tunable, and efficient approach for the HTS of anti-cancer drugs compared to conventional 2D systems.
Collapse
Affiliation(s)
- Atena Malakpour-Permlid
- Center for Intelligent Drug Delivery and Sensing Using Microcontainers and Nanomechanics (IDUN), Department of Health Technology, Technical University of Denmark, Kongens Lyngby 2800, Denmark
| | - Manuel Marcos Rodriguez
- Center for Intelligent Drug Delivery and Sensing Using Microcontainers and Nanomechanics (IDUN), Department of Health Technology, Technical University of Denmark, Kongens Lyngby 2800, Denmark
| | - Gavrielle R. Untracht
- Department of Health Technology, Technical University of Denmark, Kongens Lyngby 2800, Denmark
| | - Peter E. Andersen
- Department of Health Technology, Technical University of Denmark, Kongens Lyngby 2800, Denmark
| | | | - Anja Boisen
- Center for Intelligent Drug Delivery and Sensing Using Microcontainers and Nanomechanics (IDUN), Department of Health Technology, Technical University of Denmark, Kongens Lyngby 2800, Denmark
| | - Kinga Zór
- Center for Intelligent Drug Delivery and Sensing Using Microcontainers and Nanomechanics (IDUN), Department of Health Technology, Technical University of Denmark, Kongens Lyngby 2800, Denmark
- BioInnovation Institute Foundation, Copenhagen N 2200, Denmark
- Innovation Acta S.r.l., Siena, Via delle 1-53100, Italy
| |
Collapse
|
2
|
Xiong T, Wang K. Reconstructing the hepatocellular carcinoma microenvironment: the current status and challenges of 3D culture technology. Discov Oncol 2025; 16:506. [PMID: 40208520 PMCID: PMC11985711 DOI: 10.1007/s12672-025-02290-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/09/2024] [Accepted: 04/01/2025] [Indexed: 04/11/2025] Open
Abstract
Hepatocellular carcinoma (HCC), with high incidence and mortality rates among digestive system diseases, has become a focal point for researchers. However, the more we learn about HCC, the more apparent it becomes that our understanding is still superficial. The successes and failures of numerous studies underscore the urgent need for precision medicine in cancer treatment. A crucial aspect of preclinical research in precision medicine is the experimental model, particularly cell culture models. Among these, 3D cell culture models can effectively integrate and simulate the tumor microenvironment, closely reflecting the in vivo conditions of patients. This capability provides a solid theoretical foundation for personalized treatment approaches. In this review, we first outline the common in vitro 3D cell culture models and examine the essential elements within the tumor microenvironment, followed by insights into the current state and future developments of 3D in vitro cell models for HCC.
Collapse
Affiliation(s)
- Ting Xiong
- Division of Hepato-Biliary-Pancreatic Surgery, Department of General Surgery, The Second Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, China
- Jiangxi Province Engineering Research Center of Hepatobiliary Disease, The Second Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, China
| | - Kai Wang
- The MOE Basic Research and Innovation Center for the Targeted Therapeutics of Solid Tumors, The Second Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, China.
- Jiangxi Provincial Clinical Research Center for General Surgery Disease, The Second Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, China.
| |
Collapse
|
3
|
Mangani S, Kremmydas S, Karamanos NK. Mimicking the Complexity of Solid Tumors: How Spheroids Could Advance Cancer Preclinical Transformative Approaches. Cancers (Basel) 2025; 17:1161. [PMID: 40227664 PMCID: PMC11987746 DOI: 10.3390/cancers17071161] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2025] [Revised: 03/27/2025] [Accepted: 03/28/2025] [Indexed: 04/15/2025] Open
Abstract
Traditional 2D cell culture models present significant limitations in replicating the intricate architecture and microenvironment of in vivo solid tumors, which are essential for accurately studying cancer initiation, growth, progression, and metastasis. This underscores the need for the development of advanced preclinical models to accelerate research outcomes. Emerging 3D cell culture systems, particularly spheroid models, provide a more realistic representation of solid tumor properties by capturing the complex interactions occurring within the tumor microenvironment, including the extracellular matrix dynamics that influence cancer progression. Among solid tumors, breast cancer remains the most frequently diagnosed cancer among women globally and a leading cause of cancer-related mortality. Here we emphasize the value of breast cancer cell-derived spheroids in revealing differential molecular characteristics and understanding cancer cell properties during the early stages of invasion into adjacent tissues. Conclusively, this study underscores the urgent need to adopt 3D cell culture platforms, given their significant contributions to advanced cancer research and pharmaceutical targeting. This may well offer a transformative approach for preclinical studies and enhance our ability to test therapeutic efficiency in conditions that closely mimic the growth and progression of in vivo solid tumors.
Collapse
Affiliation(s)
| | | | - Nikos K. Karamanos
- Biochemistry, Biochemical Analysis & Matrix Pathobiology Research Group, Laboratory of Biochemistry, Department of Chemistry, University of Patras, 26504 Patras, Greece
| |
Collapse
|
4
|
Lo Cicero A, Campora S, Lo Buglio G, Cinà P, Lo Pinto M, Scilabra SD, Ghersi G. Enhancing therapeutic efficacy through degradation of endogenous extracellular matrix in primary breast tumor spheroids. FEBS J 2025. [PMID: 40098313 DOI: 10.1111/febs.70069] [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: 07/18/2024] [Revised: 11/08/2024] [Accepted: 03/10/2025] [Indexed: 03/19/2025]
Abstract
Solid tumors have a complex extracellular matrix (ECM) that significantly affects tumor behavior and response to therapy. Understanding the ECM's role is crucial for advancing cancer research and treatment. This study established an in vitro model using primary cells isolated from a rat breast tumor to generate three-dimensional spheroids. Monolayer cells and spheroid cultures exhibited different protein expression patterns, with primary tumor spheroids presenting an increased level of ECM-related proteins and a more complex extracellular environment. Furthermore, spheroids produce endogenous collagen type I matrix, which is the main component of the tumoral ECM. This matrix is arranged predominantly around the 3D structure, mimicking the conditions of solid tumors. Treatments with recombinant collagenases class II (acting on the linear collagen region) and class I (acting on the 3D-helix region) completely degrade collagen within the spheroid structure. Collagenase pretreatment enhances the accessibility of the anticancer drug doxorubicin to penetrate the core of spheroids and sensitize them to doxorubicin-induced cytotoxicity. Our findings highlight the importance of overcoming drug resistance in breast cancer by targeting the ECM and proposing a novel strategy for improving therapeutic outcomes in solid tumors. By employing a three-dimensional spheroid model, with an endogenous ECM, we can offer more relevant insights into tumor biology and treatment responses.
Collapse
Affiliation(s)
- Alessandra Lo Cicero
- Department of Biological, Chemical and Pharmaceutical Sciences and Technologies (STEBICEF), University of Palermo, Italy
| | - Simona Campora
- Department of Biological, Chemical and Pharmaceutical Sciences and Technologies (STEBICEF), University of Palermo, Italy
- Department of Biomedical Engineering Bioscience Center of the University of Cincinnati, OH, USA
| | - Gabriele Lo Buglio
- Department of Biological, Chemical and Pharmaceutical Sciences and Technologies (STEBICEF), University of Palermo, Italy
- Department of Pharmacy, University of Copenhagen, Denmark
| | | | - Margot Lo Pinto
- Proteomics Group of Ri.MED Foundation, Research Department IRCCS ISMETT (Istituto Mediterraneo per i Trapianti e Terapie ad Alta Specializzazione), Palermo, Italy
| | - Simone Dario Scilabra
- Proteomics Group of Ri.MED Foundation, Research Department IRCCS ISMETT (Istituto Mediterraneo per i Trapianti e Terapie ad Alta Specializzazione), Palermo, Italy
| | - Giulio Ghersi
- Department of Biological, Chemical and Pharmaceutical Sciences and Technologies (STEBICEF), University of Palermo, Italy
- Abiel Srl, Palermo, Italy
| |
Collapse
|
5
|
Duong VT, Dang TT, Le VP, Le TH, Nguyen CT, Phan HL, Seo J, Lin CC, Back SH, Koo KI. Direct extrusion of multifascicle prevascularized human skeletal muscle for volumetric muscle loss surgery. Biomaterials 2025; 314:122840. [PMID: 39321685 DOI: 10.1016/j.biomaterials.2024.122840] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2024] [Revised: 09/12/2024] [Accepted: 09/13/2024] [Indexed: 09/27/2024]
Abstract
Skeletal muscle is composed of multiple fascicles, which are parallel bundles of muscle fibers surrounded by connective tissues that contain blood vessels and nerves. Here, we fabricated multifascicle human skeletal muscle scaffolds that mimic the natural structure of human skeletal muscle bundles using a seven-barrel nozzle. For the core material to form the fascicle structure, human skeletal myoblasts were encapsulated in Matrigel with calcium chloride. Meanwhile, the shell that plays a role as the connective tissue, human fibroblasts and human umbilical vein endothelial cells within a mixture of porcine muscle decellularized extracellular matrix and sodium alginate at a 95:5 ratio was used. We assessed four types of extruded scaffolds monolithic-monoculture (Mo-M), monolithic-coculture (Mo-C), multifascicle-monoculture (Mu-M), and multifascicle-coculture (Mu-C) to determine the structural effect of muscle mimicking scaffold. The Mu-C scaffold outperformed other scaffolds in cell proliferation, differentiation, vascularization, mechanical properties, and functionality. In an in vivo mouse model of volumetric muscle loss, the Mu-C scaffold effectively regenerated the tibialis anterior muscle defect, demonstrating its potential for volumetric muscle transplantation. Our nozzle will be further used to produce other volumetric functional tissues, such as tendons and peripheral nerves.
Collapse
Affiliation(s)
- Van Thuy Duong
- Department of Electrical, Electronic and Computer Engineering, University of Ulsan, Ulsan, 44610, Republic of Korea; Weldon School of Biomedical Engineering, Purdue University, West Lafayette, IN 47907, USA.
| | - Thao Thi Dang
- School of Biological Sciences, University of Ulsan, Ulsan, 44610, Republic of Korea; Department of Medical and Molecular Genetics, Indiana University School of Medicine, Indianapolis, IN 46202, USA.
| | - Van Phu Le
- Department of Electrical, Electronic and Computer Engineering, University of Ulsan, Ulsan, 44610, Republic of Korea.
| | - Thi Huong Le
- Department of Electrical, Electronic and Computer Engineering, University of Ulsan, Ulsan, 44610, Republic of Korea.
| | - Chanh Trung Nguyen
- Department of Electrical, Electronic and Computer Engineering, University of Ulsan, Ulsan, 44610, Republic of Korea.
| | - Huu Lam Phan
- Department of Electrical, Electronic and Computer Engineering, University of Ulsan, Ulsan, 44610, Republic of Korea.
| | - Jongmo Seo
- Department of Electrical and Computer Engineering, Seoul National University, Seoul, 08826, Republic of Korea; Seoul National University Hospital Biomedical Research Institute, Seoul, 03080, Republic of Korea.
| | - Chien-Chi Lin
- Weldon School of Biomedical Engineering, Purdue University, West Lafayette, IN 47907, USA.
| | - Sung Hoon Back
- School of Biological Sciences, University of Ulsan, Ulsan, 44610, Republic of Korea; Basic-Clinical Convergence Research Institute, University of Ulsan, Ulsan, Republic of Korea.
| | - Kyo-In Koo
- Department of Electrical, Electronic and Computer Engineering, University of Ulsan, Ulsan, 44610, Republic of Korea; Basic-Clinical Convergence Research Institute, University of Ulsan, Ulsan, Republic of Korea.
| |
Collapse
|
6
|
Padmanaban AM, Ganesan K, Ramkumar KM. A Co-Culture System for Studying Cellular Interactions in Vascular Disease. Bioengineering (Basel) 2024; 11:1090. [PMID: 39593750 PMCID: PMC11591305 DOI: 10.3390/bioengineering11111090] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2024] [Revised: 10/17/2024] [Accepted: 10/21/2024] [Indexed: 11/28/2024] Open
Abstract
Cardiovascular diseases (CVDs) are leading causes of morbidity and mortality globally, characterized by complications such as heart failure, atherosclerosis, and coronary artery disease. The vascular endothelium, forming the inner lining of blood vessels, plays a pivotal role in maintaining vascular homeostasis. The dysfunction of endothelial cells contributes significantly to the progression of CVDs, particularly through impaired cellular communication and paracrine signaling with other cell types, such as smooth muscle cells and macrophages. In recent years, co-culture systems have emerged as advanced in vitro models for investigating these interactions and mimicking the pathological environment of CVDs. This review provides an in-depth analysis of co-culture models that explore endothelial cell dysfunction and the role of cellular interactions in the development of vascular diseases. It summarizes recent advancements in multicellular co-culture models, their physiological and therapeutic relevance, and the insights they provide into the molecular mechanisms underlying CVDs. Additionally, we evaluate the advantages and limitations of these models, offering perspectives on how they can be utilized for the development of novel therapeutic strategies and drug testing in cardiovascular research.
Collapse
Affiliation(s)
- Abirami M. Padmanaban
- Department of Biotechnology, School of Bioengineering, SRM Institute of Science and Technology, Kattankulathur 603 203, Tamil Nadu, India;
| | - Kumar Ganesan
- School of Chinese Medicine, Li Ka Shing Faculty of Medicine, The University of Hong Kong, 10 Sassoon Road, Pokfulam, Hong Kong 999077, China;
| | - Kunka Mohanram Ramkumar
- Department of Biotechnology, School of Bioengineering, SRM Institute of Science and Technology, Kattankulathur 603 203, Tamil Nadu, India;
| |
Collapse
|
7
|
Okamoto N, Taniura N, Nakayama T, Tanaka E, Kageyama Y, Noujima M, Kushima R, Mukaisho KI. Three-Dimensional Culture of Glioblastoma Cells Using a Tissueoid Cell Culture System. Acta Histochem Cytochem 2024; 57:149-155. [PMID: 39552931 PMCID: PMC11565224 DOI: 10.1267/ahc.24-00043] [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: 08/06/2024] [Accepted: 08/12/2024] [Indexed: 11/19/2024] Open
Abstract
In classical cell culture techniques, cancer cells typically proliferate in a single layer by adhering to the undersurface of laboratory vessels. Consequently, concerns have been raised regarding the fidelity of the morphological and functional characteristics of these cultured cancer cells compared to those of their in vivo counterparts. Our previous studies have investigated various epithelial malignant tumors utilizing the Tissueoid cell culture system, a three-dimensional (3D) cultivation method employing Cellbed-a nonwoven sheet composed of high-purity silica fibers as a scaffold. In this investigation, we have achieved successful 3D culturing of glioblastoma cells (A172 and T98G), which are non-epithelial in nature. As such our focus is to juxtapose their morphological features against that of those cultivated via conventional two-dimensional (2D) methods. Our findings will be elucidated using immunostaining, immunofluorescence staining, and scanning electron microscopy, substantiated with accompanying imaging. Notably, cells cultured in the 3D environment exhibited distinct morphological attributes compared to those of their 2D counterparts, notably featuring pronounced cellular protrusions. We envisage the continued utilization of the 3D culture platform to facilitate diverse avenues of research, encompassing the exploration of novel therapeutic modalities for glioblastoma cells and beyond.
Collapse
Affiliation(s)
- Natsume Okamoto
- Division of Human Pathology, Department of Pathology, Shiga University of Medical Science, Otsu, Japan
| | - Naoko Taniura
- Division of Human Pathology, Department of Pathology, Shiga University of Medical Science, Otsu, Japan
- Education Center for Medicine and Nursing, Shiga University of Medical Science, Otsu, Japan
| | - Takahisa Nakayama
- Division of Human Pathology, Department of Pathology, Shiga University of Medical Science, Otsu, Japan
| | - Eri Tanaka
- Division of Human Pathology, Department of Pathology, Shiga University of Medical Science, Otsu, Japan
| | - Yusuke Kageyama
- Division of Human Pathology, Department of Pathology, Shiga University of Medical Science, Otsu, Japan
| | - Mai Noujima
- Division of Human Pathology, Department of Pathology, Shiga University of Medical Science, Otsu, Japan
| | - Ryoji Kushima
- Division of Human Pathology, Department of Pathology, Shiga University of Medical Science, Otsu, Japan
| | - Ken-ichi Mukaisho
- Education Center for Medicine and Nursing, Shiga University of Medical Science, Otsu, Japan
| |
Collapse
|
8
|
Hu W, Cao M, Liao L, Liao Y, He Y, Ma M, Wang S, Guan Y. An Automated Digital Microfluidic System Based on Inkjet Printing. MICROMACHINES 2024; 15:1285. [PMID: 39597098 PMCID: PMC11596180 DOI: 10.3390/mi15111285] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/22/2024] [Revised: 10/16/2024] [Accepted: 10/20/2024] [Indexed: 11/29/2024]
Abstract
Cellular interactions, such as intercellular communication and signal transduction, can be enhanced within three-dimensional cell spheroids, contributing significantly to cellular viability and proliferation. This is crucial for advancements in cancer research, drug testing, and personalized medicine. The dimensions of the cell spheroids play a pivotal role in their functionality, affecting cell proliferation and differentiation, intercellular interactions, gene expression, protein synthesis, drug penetration, and metabolism. Consequently, different spheroid sizes may be required for various drug sensitivity experiments. However, conventional 3D cell spheroid cultures suffer from challenges such as size inconsistency, poor uniformity, and low throughput. To address these issues, we have developed an automated, intelligent system based on inkjet printing. This system allows for precise control of droplet volume by adjusting algorithms, thereby enabling the formation of spheroids of varying sizes. For spheroids of a single size, the printing pattern can be modified to achieve a coefficient of variation within 10% through a bidirectional compensation method. Furthermore, the system is equipped with an automatic pipetting module, which facilitates the high-throughput preparation of cell spheroids. We have implemented a 3 × 3 spheroid array in a 24-well plate, printing a total of 216 spheroids in just 11 min. Last, we attempted to print mouse small intestinal organoids and cultured them for 7 days, followed by immunofluorescent staining experiments. The results indicate that our equipment is capable of supporting the culture of organoids, which is of great significance for high-throughput drug screening and personalized medicine.
Collapse
Affiliation(s)
- Wansheng Hu
- School of Microelectronics, Shanghai University, Shanghai 201800, China;
| | - Ming Cao
- Shanghai Aure Technology Limited Company, Shanghai 201800, China
| | - Lingni Liao
- Shanghai Aure Technology Limited Company, Shanghai 201800, China
| | - Yuanhong Liao
- Shanghai Aure Technology Limited Company, Shanghai 201800, China
| | - Yuhan He
- Shanghai Aure Technology Limited Company, Shanghai 201800, China
| | - Mengxiao Ma
- Shanghai Aure Technology Limited Company, Shanghai 201800, China
| | - Simao Wang
- Shanghai Aure Technology Limited Company, Shanghai 201800, China
| | - Yimin Guan
- School of Microelectronics, Shanghai University, Shanghai 201800, China;
- Shanghai Aure Technology Limited Company, Shanghai 201800, China
| |
Collapse
|
9
|
Moro-López M, Farré R, Otero J, Sunyer R. Trusting the forces of our cell lines. Cells Dev 2024; 179:203931. [PMID: 38852676 DOI: 10.1016/j.cdev.2024.203931] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/29/2024] [Revised: 05/03/2024] [Accepted: 06/04/2024] [Indexed: 06/11/2024]
Abstract
Cells isolated from their native tissues and cultured in vitro face different selection pressures than those cultured in vivo. These pressures induce a profound transformation that reshapes the cell, alters its genome, and transforms the way it senses and generates forces. In this perspective, we focus on the evidence that cells cultured on conventional polystyrene substrates display a fundamentally different mechanobiology than their in vivo counterparts. We explore the role of adhesion reinforcement in this transformation and to what extent it is reversible. We argue that this mechanoadaptation is often understood as a mechanical memory. We propose some strategies to mitigate the effects of on-plastic culture on mechanobiology, such as organoid-inspired protocols or mechanical priming. While isolating cells from their native tissues and culturing them on artificial substrates has revolutionized biomedical research, it has also transformed cellular forces. Only by understanding and controlling them, we can improve their truthfulness and validity.
Collapse
Affiliation(s)
- Marina Moro-López
- Unit of Biophysics and Bioengineering, School of Medicine and Health Sciences, University of Barcelona, Barcelona, Spain
| | - Ramon Farré
- Unit of Biophysics and Bioengineering, School of Medicine and Health Sciences, University of Barcelona, Barcelona, Spain; Centro de Investigación Biomédica en Red de Enfermedades Respiratorias (CIBER-RES), Barcelona, Spain; Institut Investigacions Biomèdiques August Pi Sunyer (IDIBAPS), Barcelona, Spain
| | - Jorge Otero
- Unit of Biophysics and Bioengineering, School of Medicine and Health Sciences, University of Barcelona, Barcelona, Spain; Centro de Investigación Biomédica en Red de Enfermedades Respiratorias (CIBER-RES), Barcelona, Spain; Institute of Nanoscience and Nanotechnology (IN2UB), University of Barcelona, Barcelona, Spain
| | - Raimon Sunyer
- Unit of Biophysics and Bioengineering, School of Medicine and Health Sciences, University of Barcelona, Barcelona, Spain; Institute of Nanoscience and Nanotechnology (IN2UB), University of Barcelona, Barcelona, Spain; Centro de Investigación Biomédica en Red de Bioingeniería (CIBER-BBN), Barcelona, Spain.
| |
Collapse
|
10
|
Bussi C, Lai R, Athanasiadi N, Gutierrez MG. Physiologic medium renders human iPSC-derived macrophages permissive for M. tuberculosis by rewiring organelle function and metabolism. mBio 2024; 15:e0035324. [PMID: 38984828 PMCID: PMC11323749 DOI: 10.1128/mbio.00353-24] [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: 02/05/2024] [Accepted: 06/04/2024] [Indexed: 07/11/2024] Open
Abstract
In vitro studies are crucial for our understanding of the human macrophage immune functions. However, traditional in vitro culture media poorly reflect the metabolic composition of blood, potentially affecting the outcomes of these studies. Here, we analyzed the impact of a physiological medium on human induced pluripotent stem cell (iPSC)-derived macrophages (iPSDM) function. Macrophages cultured in a human plasma-like medium (HPLM) were more permissive to Mycobacterium tuberculosis (Mtb) replication and showed decreased lipid metabolism with increased metabolic polarization. Functionally, we discovered that HPLM-differentiated macrophages showed different metabolic organelle content and activity. Specifically, HPLM-differentiated macrophages displayed reduced lipid droplet and peroxisome content, increased lysosomal proteolytic activity, and increased mitochondrial activity and dynamics. Inhibiting or inducing lipid droplet formation revealed that lipid droplet content is a key factor influencing macrophage permissiveness to Mtb. These findings underscore the importance of using physiologically relevant media in vitro for accurately studying human macrophage function. IMPORTANCE This work compellingly demonstrates that the choice of culture medium significantly influences M. tuberculosis replication outcomes, thus emphasizing the importance of employing physiologically relevant media for accurate in vitro host-pathogen interaction studies. We anticipate that our work will set a precedent for future research with clinical relevance, particularly in evaluating antibiotic efficacy and resistance in cellulo.
Collapse
Affiliation(s)
- Claudio Bussi
- The Francis Crick Institute, London, United Kingdom
- School of Biological Sciences, Nanyang Technological University, Singapore, Singapore
| | - Rachel Lai
- The Francis Crick Institute, London, United Kingdom
- Department of Infectious Diseases, Imperial College London, London, United Kingdom
| | | | | |
Collapse
|
11
|
Mastronikolis NS, Kyrodimos E, Piperigkou Z, Spyropoulou D, Delides A, Giotakis E, Alexopoulou M, Bakalis NA, Karamanos NK. Matrix-based molecular mechanisms, targeting and diagnostics in oral squamous cell carcinoma. IUBMB Life 2024; 76:368-382. [PMID: 38168122 DOI: 10.1002/iub.2803] [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: 08/22/2023] [Accepted: 11/22/2023] [Indexed: 01/05/2024]
Abstract
Oral squamous cell carcinoma (OSCC) is a head and neck cancer (HNC) with a high mortality rate. OSCC is developed in the oral cavity and it is triggered by many etiologic factors and can metastasize both regionally and distantly. Recent research advances in OSCC improved our understanding on the molecular mechanisms involved in and the initiation of OSCC metastasis. The key roles of the extracellular matrix (ECM) in OSCC are an emerging area of intensive research as the ECM macromolecular network is actively involved in events that regulate cellular morphological and functional properties, transcription and cell signaling mechanisms in invasion and metastasis. The provisional matrix that is formed by cancer cells is profoundly different in composition and functions as compared with the matrix of normal tissue. Fibroblasts are mainly responsible for matrix production and remodeling, but in cancer, the tumor matrix in the tumor microenvironment (TME) also originates from cancer cells. Even though extensive research has been conducted on the role of ECM in regulating cancer pathogenesis, its role in modulating OSCC is less elucidated since there are several issues yet to be fully understood. This critical review is focused on recent research as to present and discuss on the involvement of ECM macromolecular effectors (i.e., proteoglycans, integrins, matrix metalloproteinases) in OSCC development and progression.
Collapse
Affiliation(s)
- Nicholas S Mastronikolis
- Department of Otorhinolaryngology - Head and Neck Surgery, School of Medicine, University of Patras, Patras, Greece
| | - Efthymios Kyrodimos
- 1st Otolaryngology Department, School of Medicine, National & Kapodistrian University of Athens, 'Ippokrateion' General Hospital, Athens, Greece
| | - Zoi Piperigkou
- Biochemistry, Biochemical Analysis & Matrix Pathobiology Research Group, Laboratory of Biochemistry, Department of Chemistry, University of Patras, Patras, Greece
- Foundation for Research and Technology - Hellas (FORTH)/Institute of Chemical Engineering Sciences (ICE-HT), Patras, Greece
| | - Despoina Spyropoulou
- Department of Radiation Oncology, School of Medicine, University of Patras, Patras, Greece
| | - Alexander Delides
- 2nd Otolaryngology Department, School of Medicine, National & Kapodistrian University of Athens, 'Attikon' University Hospital, Athens, Greece
| | - Evangelos Giotakis
- 1st Otolaryngology Department, School of Medicine, National & Kapodistrian University of Athens, 'Ippokrateion' General Hospital, Athens, Greece
- Department of Radiation Oncology, School of Medicine, University of Patras, Patras, Greece
- 2nd Otolaryngology Department, School of Medicine, National & Kapodistrian University of Athens, 'Attikon' University Hospital, Athens, Greece
| | - Miranda Alexopoulou
- Department of Maxillofacial Surgery, University Hospital of Patras, Patras, Greece
| | - Nick A Bakalis
- Department of Nursing, University of Patras, Patras, Greece
| | - Nikos K Karamanos
- Biochemistry, Biochemical Analysis & Matrix Pathobiology Research Group, Laboratory of Biochemistry, Department of Chemistry, University of Patras, Patras, Greece
- Foundation for Research and Technology - Hellas (FORTH)/Institute of Chemical Engineering Sciences (ICE-HT), Patras, Greece
| |
Collapse
|
12
|
Jamshidi N, Jamshidi N, Modarresi Chahardehi A, Shams E, Chaleshi V. A promising breakthrough in pancreatic cancer research: The potential of spheroids as 3D models. BIOIMPACTS : BI 2024; 15:30241. [PMID: 39963557 PMCID: PMC11830132 DOI: 10.34172/bi.30241] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 12/27/2023] [Revised: 01/29/2024] [Accepted: 02/06/2024] [Indexed: 02/20/2025]
Abstract
Pancreatic ductal adenocarcinoma (PDAC) stands as the fourth leading cause of cancer-related deaths, primarily attributable to its resistance to chemotherapy, resulting in a nearly universal fatality rate. Despite the promise exhibited by numerous drugs in preclinical studies, their subsequent failure in clinical trials underscores the inherent limitations of conventional two-dimensional cell culture models commonly employed in early drug screening endeavors. The inadequacies of two-dimensional (2D) models prompted the exploration of three-dimensional (3D) culture systems, which more faithfully recapitulate the native tumor microenvironment. These 3D systems have distinct advantages over 2D models in morphology, proliferation, drug response, and protein expression. Among these 3D platforms, tumor organoids and spheroids, generated through different methodologies, have emerged as next-generation models that closely mirror aspects of pancreatic tumor biology. This comprehensive review scrutinizes pancreatic cancer spheroids' techniques, tissue sources, and applications, offering a nuanced analysis of their advantages and limitations. By comparing these distinct 3D culture systems, researchers gain valuable insights to inform the selection of optimal model designs aligned with their specific experimental objectives. The utilization of these advanced models holds significant promise for enhancing the clinical relevance of both in vitro and in vivo cancer research, thereby contributing to the development of improved therapeutics against pancreatic cancer.
Collapse
Affiliation(s)
- Nazanin Jamshidi
- Kimia Andisheh Teb Medical and Molecular Laboratory Research Co, Tehran, Iran
| | - Negar Jamshidi
- Kimia Andisheh Teb Medical and Molecular Laboratory Research Co, Tehran, Iran
| | | | - Elahe Shams
- Basic and Molecular Epidemiology of Gastrointestinal Disorders Research Center, Research Institute for Gastroenterology and Liver Diseases, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Vahid Chaleshi
- Basic and Molecular Epidemiology of Gastrointestinal Disorders Research Center, Research Institute for Gastroenterology and Liver Diseases, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| |
Collapse
|
13
|
Franchi M, Piperigkou Z, Mastronikolis NS, Karamanos N. Extracellular matrix biomechanical roles and adaptation in health and disease. FEBS J 2024; 291:430-440. [PMID: 37612040 DOI: 10.1111/febs.16938] [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: 06/08/2023] [Revised: 07/24/2023] [Accepted: 08/22/2023] [Indexed: 08/25/2023]
Abstract
Extracellular matrices (ECMs) are dynamic 3D macromolecular networks that exhibit structural characteristics and composition specific to different tissues, serving various biomechanical and regulatory functions. The interactions between ECM macromolecules such as collagen, elastin, glycosaminoglycans (GAGs), proteoglycans (PGs), fibronectin, and laminin, along with matrix effectors and water, contribute to the unique cellular and tissue functional properties during organ development, tissue homoeostasis, remodeling, disease development, and progression. Cells adapt to environmental changes by adjusting the composition and array of ECM components. ECMs, forming the 3D bioscaffolds of our body, provide mechanical support for tissues and organs and respond to the environmental variables influencing growth and final adult body shape in mammals. Different cell types display distinct adaptations to the respective ECM environments. ECMs regulate biological processes by controlling the diffusion of infections and inflammations, sensing and adapting to external stimuli and gravity from the surrounding habitat, and, in the context of cancer, interplaying with and regulating cancer cell invasion and drug resistance. Alterations in the ECM composition in pathological conditions drive adaptive responses of cells and could therefore result in abnormal cell behavior and tissue dysfunction. Understanding the biomechanical functionality, adaptation, and roles of distinct ECMs is essential for research on various pathologies, including cancer progression and multidrug resistance, which is of crucial importance for developing targeted therapies. In this Viewpoint article, we critically present and discuss specific biomechanical functions of ECMs and regulatory adaptation mechanisms in both health and disease, with a particular focus on cancer progression.
Collapse
Affiliation(s)
- Marco Franchi
- Department for Life Quality Studies, University of Bologna, Rimini, Italy
| | - Zoi Piperigkou
- Department of Chemistry, Biochemistry, Biochemical Analysis and Matrix Pathobiology Res. Group, Laboratory of Biochemistry, University of Patras, Greece
- Foundation for Research and Technology-Hellas (FORTH)/Institute of Chemical Engineering Sciences (ICE-HT), Patras, Greece
| | - Nicholas S Mastronikolis
- Department of Otorhinolaryngology-Head and Neck Surgery, School of Medicine, University of Patras, Greece
| | - Nikos Karamanos
- Department of Chemistry, Biochemistry, Biochemical Analysis and Matrix Pathobiology Res. Group, Laboratory of Biochemistry, University of Patras, Greece
- Foundation for Research and Technology-Hellas (FORTH)/Institute of Chemical Engineering Sciences (ICE-HT), Patras, Greece
| |
Collapse
|
14
|
Anameriç A, Czerwonka A, Nees M. Optimization of a Three-Dimensional Culturing Method for Assessing the Impact of Cisplatin on Notch Signaling in Head and Neck Squamous Cell Carcinoma (HNSCC). Cancers (Basel) 2023; 15:5320. [PMID: 38001580 PMCID: PMC10670464 DOI: 10.3390/cancers15225320] [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: 09/23/2023] [Revised: 11/01/2023] [Accepted: 11/02/2023] [Indexed: 11/26/2023] Open
Abstract
Head and neck squamous cell carcinoma (HNSCC) is a prevalent cancer type, with cisplatin being a primary treatment approach. However, drug resistance and therapy failure pose a significant challenge, affecting nearly 50% of patients over time. This research had two aims: (1) to optimize a 3D cell-culture method for assessing the interplay between tumor cells and cancer-associated fibroblasts (CAFs) in vitro; and (2) to study how cisplatin impacts the Notch pathway, particularly considering the role of CAFs. Using our optimized "3D sheet model" approach, we tested two HNSCC cell lines with different cisplatin sensitivities and moderate, non-mutated NOTCH1 and -3 expressions. Combining cisplatin with a γ-secretase inhibitor (crenigacestat) increased sensitivity and induced cell death in the less sensitive cell line, while cisplatin alone was more effective in the moderately sensitive line and sensitivity decreased with the Notch inhibitor. Cisplatin boosted the expression of core Notch signaling proteins in 3D monocultures of both lines, which was counteracted by crenigacestat. In contrast, the presence of patient-derived CAFs mitigated effects and protected both cell lines from cisplatin toxicity. Elevated NOTCH1 and NOTCH3 protein levels were consistently correlated with reduced cisplatin sensitivity and increased cell survival. Additionally, the Notch ligand JAG2 had additional, protective effects reducing cell death from cisplatin exposure. In summary, we observed an inverse relationship between NOTCH1 and NOTCH3 levels and cisplatin responsiveness, overall protective effects by CAFs, and a potential link between JAG2 expression with tumor cell survival.
Collapse
Affiliation(s)
| | | | - Matthias Nees
- Department of Biochemistry and Molecular Biology, Medical University of Lublin, 20-093 Lublin, Poland; (A.A.); (A.C.)
| |
Collapse
|
15
|
Piperigkou Z, Bainantzou D, Makri N, Papachristou E, Mantsou A, Choli-Papadopoulou T, Theocharis AD, Karamanos NK. Enhancement of mesenchymal stem cells' chondrogenic potential by type II collagen-based bioscaffolds. Mol Biol Rep 2023; 50:5125-5135. [PMID: 37118382 PMCID: PMC10209287 DOI: 10.1007/s11033-023-08461-x] [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: 03/13/2023] [Accepted: 04/12/2023] [Indexed: 04/30/2023]
Abstract
BACKGROUND Osteoarthritis (OA) is a common degenerative chronic disease accounting for physical pain, tissue stiffness and mobility restriction. Current therapeutic approaches fail to prevent the progression of the disease considering the limited knowledge on OA pathobiology. During OA progression, the extracellular matrix (ECM) of the cartilage is aberrantly remodeled by chondrocytes. Chondrocytes, being the main cell population of the cartilage, participate in cartilage regeneration process. To this end, modern tissue engineering strategies involve the recruitment of mesenchymal stem cells (MSCs) due to their regenerative capacity as to promote chondrocyte self-regeneration. METHODS AND RESULTS In the present study, we evaluated the role of type II collagen, as the main matrix macromolecule in the cartilage matrix, to promote chondrogenic differentiation in two MSC in vitro culture systems. The chondrogenic differentiation of human Wharton's jelly- and dental pulp-derived MSCs was investigated over a 24-day culture period on type II collagen coating to improve the binding affinity of MSCs. Functional assays, demonstrated that type II collagen promoted chondrogenic differentiation in both MSCs tested, which was confirmed through gene and protein analysis of major chondrogenic markers. CONCLUSIONS Our data support that type II collagen contributes as a natural bioscaffold enhancing chondrogenesis in both MSC models, thus enhancing the commitment of MSC-based therapeutic approaches in regenerative medicine to target OA and bring therapy closer to the clinical use.
Collapse
Affiliation(s)
- Zoi Piperigkou
- Biochemistry, Biochemical Analysis and Matrix Pathobiology Research Group, Laboratory of Biochemistry, Department of Chemistry, University of Patras, Patras, Greece
- Foundation for Research and Technology-Hellas (FORTH)/Institute of Chemical Engineering Sciences (ICE-HT), Patras, Greece
| | - Dimitra Bainantzou
- Biochemistry, Biochemical Analysis and Matrix Pathobiology Research Group, Laboratory of Biochemistry, Department of Chemistry, University of Patras, Patras, Greece
| | - Nadia Makri
- Biochemistry, Biochemical Analysis and Matrix Pathobiology Research Group, Laboratory of Biochemistry, Department of Chemistry, University of Patras, Patras, Greece
| | - Eleni Papachristou
- Laboratory of Biochemistry, Department of Chemistry, Aristotle University of Thessaloniki, Thessaloniki, Greece
| | - Aglaia Mantsou
- Laboratory of Biochemistry, Department of Chemistry, Aristotle University of Thessaloniki, Thessaloniki, Greece
| | - Theodora Choli-Papadopoulou
- Laboratory of Biochemistry, Department of Chemistry, Aristotle University of Thessaloniki, Thessaloniki, Greece
| | - Achilleas D Theocharis
- Biochemistry, Biochemical Analysis and Matrix Pathobiology Research Group, Laboratory of Biochemistry, Department of Chemistry, University of Patras, Patras, Greece.
| | - Nikos K Karamanos
- Biochemistry, Biochemical Analysis and Matrix Pathobiology Research Group, Laboratory of Biochemistry, Department of Chemistry, University of Patras, Patras, Greece.
- Foundation for Research and Technology-Hellas (FORTH)/Institute of Chemical Engineering Sciences (ICE-HT), Patras, Greece.
| |
Collapse
|
16
|
Badve SS, Gökmen-Polar Y. Targeting the Tumor-Tumor Microenvironment Crosstalk. Expert Opin Ther Targets 2023; 27:447-457. [PMID: 37395003 DOI: 10.1080/14728222.2023.2230362] [Citation(s) in RCA: 17] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2023] [Accepted: 06/23/2023] [Indexed: 07/04/2023]
Abstract
INTRODUCTION Cancer development and progression is a complex process influenced by co-evolution of the cancer cells and their microenvironment. However, traditional anti-cancer therapy is mostly targeted toward cancer cells. To improve the efficacy of cancer drugs, the complex interactions between the tumor (T) and the tumor microenvironment (TME) should be considered while developing therapeutics. AREAS COVERED The present review article will discuss the components of T-TME as well as the potential to co-target these two distinct elements. We document that these approaches have resulted in success in preventing tumor progression and metastasis, albeit in animal models in some cases. Lastly, it is important to consider the tissue context and tumor type as these could significantly modify the role of these molecules/pathways and hence the overall likelihood of response. Furthermore, we discuss the potential strategies to target the components of tumor microenvironment in anti-cancer therapy. PubMed and ClinicalTrials.gov was searched through May 2023. EXPERT OPINION The tumor-tumor microenvironment cross talk and heterogeneity are major mechanisms conferring resistance to standard of care. Better understanding of the tissue specific T-TME interactions and dual targeting has the promise of improving cancer control and clinical outcomes.
Collapse
Affiliation(s)
- Sunil S Badve
- Department of Pathology and Laboratory Medicine, Emory University, Atlanta, GA, USA
| | - Yesim Gökmen-Polar
- Department of Pathology and Laboratory Medicine, Emory University, Atlanta, GA, USA
| |
Collapse
|