1
|
Hazari MA, Kannan G, Dasgupta S, Pavan MK, Jha AK, Sultana F, Pujahari SR, Singh S, Dutta S, Pydi SP, Dutta S, Zafar H, Bhaumik P, Kumar A, Sen S. Faster Amylin Aggregation on Fibrillar Collagen I Hastens Diabetic Progression through β-Cell Death and Loss of Function. J Am Chem Soc 2025; 147:15985-16006. [PMID: 40300850 DOI: 10.1021/jacs.4c15698] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/01/2025]
Abstract
Amyloid deposition of the neuroendocrine peptide amylin in islet tissues is a hallmark of type 2 diabetes (T2DM), leading to β-cell toxicity through nutrient deprivation, membrane rupture, and apoptosis. Though accumulation of toxic amylin aggregates in islet matrices is well documented, the role of the islet extracellular matrix in mediating amylin aggregation and its pathological consequences remains elusive. Here, we address this question by probing amylin interaction with collagen I (Col)─whose expression in the islet tissue increases during diabetes progression. By combining multiple biophysical techniques, we show that hydrophobic, hydrophilic, and cation-π interactions regulate amylin binding to Col, with fibrillar Col driving faster amylin aggregation. Amylin-entangled Col matrices containing high amounts of amylin induce death and loss of function in INS1E β-cells. Together, our results illustrate how amylin incorporation in islet matrices through amylin-Col interactions drives T2DM progression by impacting β-cell viability and insulin secretion.
Collapse
Affiliation(s)
| | - Gautam Kannan
- Department of Biosciences & Bioengineering, IIT Bombay, Mumbai 400076, India
| | - Subrata Dasgupta
- Department of Biosciences & Bioengineering, IIT Bombay, Mumbai 400076, India
| | - Musale Krushna Pavan
- Department of Computer Science and Engineering, IIT Kanpur, Kanpur 208016, India
| | - Akash Kumar Jha
- Department of Biosciences & Bioengineering, IIT Bombay, Mumbai 400076, India
| | - Farhin Sultana
- Department of Oncogene Regulation, CNCI, Kolkata 700026, India
| | | | - Simran Singh
- Department of Biological Sciences and Bioengineering, IIT Kanpur, Kanpur 208016, India
- Mehta Family Centre for Engineering in Medicine, IIT Kanpur, Kanpur 208016, India
| | - Sarbajeet Dutta
- Department of Biosciences & Bioengineering, IIT Bombay, Mumbai 400076, India
| | - Sai Prasad Pydi
- Department of Biological Sciences and Bioengineering, IIT Kanpur, Kanpur 208016, India
- Mehta Family Centre for Engineering in Medicine, IIT Kanpur, Kanpur 208016, India
| | | | - Hamim Zafar
- Department of Biological Sciences and Bioengineering, IIT Kanpur, Kanpur 208016, India
- Department of Computer Science and Engineering, IIT Kanpur, Kanpur 208016, India
- Mehta Family Centre for Engineering in Medicine, IIT Kanpur, Kanpur 208016, India
| | - Prasenjit Bhaumik
- Department of Biosciences & Bioengineering, IIT Bombay, Mumbai 400076, India
| | - Ashutosh Kumar
- Department of Biosciences & Bioengineering, IIT Bombay, Mumbai 400076, India
| | - Shamik Sen
- Department of Biosciences & Bioengineering, IIT Bombay, Mumbai 400076, India
| |
Collapse
|
2
|
Gantert B, Karakaya E, Hofmann F, Jungst T, Meinel L, Bosserhoff AK, Detsch R, Lühmann T. Alginate-Dialdehyde-Based Reporter Ink Enabling Online Detection of Matrix Metalloproteinase Activity of Encapsulated Cells. ACS Biomater Sci Eng 2025; 11:2435-2447. [PMID: 40052617 DOI: 10.1021/acsbiomaterials.4c02399] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/15/2025]
Abstract
Biofabrication and three-dimensional (3D) bioprinting enable precise spatial arrangement of cells within biomaterial scaffolds. We developed an alginate-based and Förster resonance energy transfer (FRET)-responsive "turn-on" reporter ink platform to enable real-time monitoring of matrix metalloproteinase (MMP) activity. Three distinct MMP-cleavable turn-on peptide reporters were synthesized and characterized for their cell-specific cleavage profiles using recombinant MMPs, cell-derived media, and different cell cultures (NIH3T3, HEK293, and MelHo). All turn-on reporters were covalently and site-specifically incorporated into alginate dialdehyde (ADA) to yield an MMP reporter ink. The ADA reporter ink with an MMP 13 turn-on reporter was responsive to all tested cell types over time within the cast bulk constructs. The ADA reporter ink material blended with gelatin had comparable print resolution and structural fidelity as observed for ADA. The extrusion-based bioprinted MelHo cell grids, measuring 2 × 2 cm2 and containing 1 × 106 cells/mL, exhibited MMP activity responses comparable to those of the casted reporter ink system, with a 3-fold increase observed at 24 h. This study introduces a versatile, FRET-based alginate bioink platform for the real-time monitoring of MMP activities, expanding the toolkit to understand cellular performance in bioprinted 3D constructs.
Collapse
Affiliation(s)
- Benedikt Gantert
- Institute of Pharmacy and Food Chemistry, University of Würzburg, Am Hubland, 97074 Würzburg, Germany
| | - Emine Karakaya
- Institute of Biomaterials, Department of Materials Science and Engineering, University of Erlangen-Nuremberg, Ulrich-Schalk-Str. 3, 91056 Erlangen, Germany
| | - Florian Hofmann
- Department for Functional Materials in Medicine and Dentistry, Institute of Functional Materials and Biofabrication, University of Würzburg and KeyLab Polymers for Medicine of the Bavarian Polymer Institute (BPI), 97070 Würzburg, Germany
| | - Tomasz Jungst
- Department for Functional Materials in Medicine and Dentistry, Institute of Functional Materials and Biofabrication, University of Würzburg and KeyLab Polymers for Medicine of the Bavarian Polymer Institute (BPI), 97070 Würzburg, Germany
| | - Lorenz Meinel
- Institute of Pharmacy and Food Chemistry, University of Würzburg, Am Hubland, 97074 Würzburg, Germany
- Helmholtz Institute for RNA-Based Infection Research (HIRI), Helmholtz Center for Infection Research (HZI), 97080 Würzburg, Germany
| | - Anja K Bosserhoff
- Institute of Biochemistry, Emil-Fischer-Zentrum, University of Erlangen-Nuremberg, Fahrstrasse 17, 91054 Erlangen, Germany
| | - Rainer Detsch
- Institute of Biomaterials, Department of Materials Science and Engineering, University of Erlangen-Nuremberg, Ulrich-Schalk-Str. 3, 91056 Erlangen, Germany
| | - Tessa Lühmann
- Institute of Pharmacy and Food Chemistry, University of Würzburg, Am Hubland, 97074 Würzburg, Germany
| |
Collapse
|
3
|
Narasimhan BN, Fraley SI. Matrix degradation enhances stress relaxation, regulating cell adhesion and spreading. Proc Natl Acad Sci U S A 2025; 122:e2416771122. [PMID: 40131951 PMCID: PMC12002262 DOI: 10.1073/pnas.2416771122] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2024] [Accepted: 02/09/2025] [Indexed: 03/27/2025] Open
Abstract
In native extracellular matrices (ECM), cells utilize matrix metalloproteinases (MMPs) to degrade and remodel their microenvironment. Accordingly, synthetic matrices have been engineered to permit MMP-mediated cleavage, facilitating cell spreading, migration, and interactions. However, the interplay between matrix degradability and mechanical properties remains underexplored. We hypothesized that MMP activity induces immediate mechanical alterations in the ECM, which are subsequently detected by cells. We observed that both fibrillar collagen and synthetic degradable matrices exhibit enhanced stress relaxation following MMP exposure. Cells responded to these variations in relaxation by modulating their spreading and focal adhesions. Furthermore, we demonstrated that stress relaxation and cell spreading can be precisely controlled through the rational design of matrix degradability. These findings establish a fundamental link between matrix degradability and stress relaxation, with potential implications for a broad spectrum of biological applications.
Collapse
|
4
|
Chen X, Zhong X, Zhang F, Zhou X, Yue X, Li X. Molecular mechanisms and therapeutic targets in glioblastoma multiforme: network and single-cell analyses. Sci Rep 2025; 15:10558. [PMID: 40148380 PMCID: PMC11950307 DOI: 10.1038/s41598-025-92867-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2024] [Accepted: 03/03/2025] [Indexed: 03/29/2025] Open
Abstract
Glioblastoma multiforme (GBM) is a highly aggressive brain tumor associated with poor survival outcomes and is driven by a complex tumor microenvironment (TME) that promotes tumor progression and treatment resistance. To explore the role of the TME in GBM, we analyzed glioma-related microarray and single-cell RNA sequencing (scRNA-seq) datasets from the Gene Expression Omnibus (GEO). Functional enrichment and weighted gene coexpression network analyses revealed distinct immune profiles, metabolic alterations, and differences in chemotherapeutic drug sensitivity between the high-risk and low-risk patient groups. scRNA-seq data processed with the 'Seurat' package were used to identify differentially expressed genes in pericytes, endothelial cells, and glioma cells, particularly those involved in extracellular matrix (ECM) remodeling. A 17-gene prognostic signature developed through Cox regression and LASSO analyses revealed that key genes (COL1A1, COL4A1, and VIM) were significantly associated with survival outcomes in GBM patients. Drug sensitivity analyses using data from the Genomics of Drug Sensitivity in Cancer (GDSC) and Cancer Therapeutics Response Portal (CTRP) identified potential targeted therapies for GBM, including SB-505,124, staurosporine, and AZD8186. This integrative study underscores the critical roles of the ECM and synaptic remodeling in GBM and suggests novel therapeutic targets to improve personalized treatment strategies for GBM patients.
Collapse
Affiliation(s)
- Xiangyu Chen
- Key Laboratory of Major Brain Disease and Aging Research (Ministry of Education), Institute for Brain Science and Disease, Chongqing Medical University, Chongqing, 400016, China
| | - Xiao Zhong
- Key Laboratory of Major Brain Disease and Aging Research (Ministry of Education), Institute for Brain Science and Disease, Chongqing Medical University, Chongqing, 400016, China
| | - Feifei Zhang
- Department of Blood Transfusion, School of Medicine, Sichuan Provincial People's Hospital, University of Electronic Science and Technology of China, Chengdu, 610021, Sichuan, China
| | - Xiaomei Zhou
- Sichuan Provincial Chengdu Second People's Hospital, Chengdu, 610021, Sichuan, China
| | - Xiaofeng Yue
- Department of Urology, The Third Affiliated Hospital of Chongqing Medical University, Chongqing, 401120, China.
| | - Xueru Li
- Key Laboratory of Major Brain Disease and Aging Research (Ministry of Education), Institute for Brain Science and Disease, Chongqing Medical University, Chongqing, 400016, China.
| |
Collapse
|
5
|
Peng Y, Huang YH, Luo X, Li MC, Xiao QQ, Qiu L, Fu Q. Magnoflorine ameliorates cartilage degradation in osteoarthritis through inhibition of mitochondrial reactive oxygen species-mediated activation of the NLRP3 inflammasome. JOURNAL OF ASIAN NATURAL PRODUCTS RESEARCH 2025:1-22. [PMID: 40035483 DOI: 10.1080/10286020.2025.2472898] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/30/2024] [Revised: 02/20/2025] [Accepted: 02/23/2025] [Indexed: 03/05/2025]
Abstract
This study investigated the role of magnoflorine (MAG) on cartilage protection in osteoarthritis. In vitro studies showed that MAG decreased the expression of inflammatory factors and inhibited extracellular matrix degradation in lipopolysaccharide- and ATP-stimulated C28/I2 cells. Importantly, MAG reduced the levels of pyroptosis-related proteins, including NLRP3, ASC, cleaved-caspase 1, GSDMD-N, IL-18, and IL-1β. Mechanistically, MAG reduced mtROS production and inhibited the activation of the NF-κB signaling pathway. In vivo study demonstrated that sodium iodoacetate-induced cartilage degradation and inflammatory factor release were reversed by MAG. Overall, MAG could inhibit mtROS-mediated NLRP3 inflammasome activation by suppressing mitochondrial dysfunction to ameliorate osteoarthritis.
Collapse
Affiliation(s)
- Yi Peng
- Sichuan Industrial Institute of Antibiotics, School of Pharmacy, Chengdu University, Chengdu 610106, China
| | - Yue-Hui Huang
- Sichuan Industrial Institute of Antibiotics, School of Pharmacy, Chengdu University, Chengdu 610106, China
| | - Xiao Luo
- Chengdu Institute for Drug Control, NMPA Key Laboratory for Quality Monitoring and Evaluation of Traditional Chinese Medicine, Chengdu 610045, China
| | - Mei-Chen Li
- Sichuan Industrial Institute of Antibiotics, School of Pharmacy, Chengdu University, Chengdu 610106, China
| | - Qing-Qing Xiao
- Sichuan Industrial Institute of Antibiotics, School of Pharmacy, Chengdu University, Chengdu 610106, China
| | - Lu Qiu
- Sichuan Industrial Institute of Antibiotics, School of Pharmacy, Chengdu University, Chengdu 610106, China
| | - Qiang Fu
- Sichuan Industrial Institute of Antibiotics, School of Pharmacy, Chengdu University, Chengdu 610106, China
| |
Collapse
|
6
|
Saha SK, Sarkar M, Srivastava M, Dutta S, Sen S. Nuclear α-actinin-4 regulates breast cancer invasiveness and EMT. Cytoskeleton (Hoboken) 2025; 82:145-157. [PMID: 39143850 DOI: 10.1002/cm.21901] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2024] [Revised: 07/12/2024] [Accepted: 07/29/2024] [Indexed: 08/16/2024]
Abstract
Epithelial-to-mesenchymal transition (EMT) is a key process where cells lose their adhesion properties and augment their invasive properties. α-Actinin4 (ACTN4) is an actin crosslinking protein that responds to mechanical stimuli and is found to be elevated in breast cancer patients. While ACTN4 has been implicated in regulating cancer invasiveness by modulating cytoskeletal organization, its nuclear functions remain much less explored. Here we address this question by first establishing a correlation between nuclear localization and invasiveness in breast cancer cells. Using cancer databases, we then establish a correlation between ACTN4 expression and EMT in breast cancer. Interestingly, TGFβ-induced EMT induction in MCF10A normal mammary epithelial cells leads to increased ACTN4 expression and nuclear enrichment. We then show that ACTN4 knockdown in MDA-MB-231 breast cancer cells, which harbor sizeable fraction of nuclear ACTN4, leads to reduced invasiveness and loss of mesenchymal traits. Similar behavior was observed in knockdown cells expressing K255E ACTN4, which is primarily localized to the cytosol. Together, our findings establish a role for nuclear ACTN4 in regulating invasiveness via modulation of EMT.
Collapse
Affiliation(s)
- Sumon Kumar Saha
- Department of Biosciences & Bioengineering, IIT Bombay, Mumbai, India
| | - Madhurima Sarkar
- Department of Biosciences & Bioengineering, IIT Bombay, Mumbai, India
| | | | - Sarbajeet Dutta
- Department of Biosciences & Bioengineering, IIT Bombay, Mumbai, India
| | - Shamik Sen
- Department of Biosciences & Bioengineering, IIT Bombay, Mumbai, India
| |
Collapse
|
7
|
Zhang S, Huang J, Jiang Z, Tong H, Ma X, Liu Y. Tumor microbiome: roles in tumor initiation, progression, and therapy. MOLECULAR BIOMEDICINE 2025; 6:9. [PMID: 39921821 PMCID: PMC11807048 DOI: 10.1186/s43556-025-00248-9] [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: 08/02/2024] [Revised: 01/06/2025] [Accepted: 01/21/2025] [Indexed: 02/10/2025] Open
Abstract
Over the past few years, the tumor microbiome is increasingly recognized for its multifaceted involvement in cancer initiation, progression, and metastasis. With the application of 16S ribosomal ribonucleic acid (16S rRNA) sequencing, the intratumoral microbiome, also referred to as tumor-intrinsic or tumor-resident microbiome, has also been found to play a significant role in the tumor microenvironment (TME). Understanding their complex functions is critical for identifying new therapeutic avenues and improving treatment outcomes. This review first summarizes the origins and composition of these microbial communities, emphasizing their adapted diversity across a diverse range of tumor types and stages. Moreover, we outline the general mechanisms by which specific microbes induce tumor initiation, including the activation of carcinogenic pathways, deoxyribonucleic acid (DNA) damage, epigenetic modifications, and chronic inflammation. We further propose the tumor microbiome may evade immunity and promote angiogenesis to support tumor progression, while uncovering specific microbial influences on each step of the metastatic cascade, such as invasion, circulation, and seeding in secondary sites. Additionally, tumor microbiome is closely associated with drug resistance and influences therapeutic efficacy by modulating immune responses, drug metabolism, and apoptotic pathways. Furthermore, we explore innovative microbe-based therapeutic strategies, such as engineered bacteria, oncolytic virotherapy, and other modalities aimed at enhancing immunotherapeutic efficacy, paving the way for microbiome-centered cancer treatment frameworks.
Collapse
Affiliation(s)
- Shengxin Zhang
- Department of Biotherapy, Cancer Center and State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, 610041, Sichuan Province, China
| | - Jing Huang
- Department of Medical Ultrasound, West China Hospital of Sichuan University, 37 Guoxue Lane, Wuhou District, Chengdu, 610041, Sichuan Province, China
| | - Zedong Jiang
- Department of Biotherapy, Cancer Center and State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, 610041, Sichuan Province, China
| | - Huan Tong
- Department of Biotherapy, Cancer Center and State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, 610041, Sichuan Province, China
| | - Xuelei Ma
- Department of Biotherapy, Cancer Center and State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, 610041, Sichuan Province, China.
| | - Yang Liu
- Day Surgery Center, General Practice Medical Center, West China Hospital, Sichuan University, Chengdu, Sichuan, 610041, P. R. China.
| |
Collapse
|
8
|
Chastney MR, Kaivola J, Leppänen VM, Ivaska J. The role and regulation of integrins in cell migration and invasion. Nat Rev Mol Cell Biol 2025; 26:147-167. [PMID: 39349749 DOI: 10.1038/s41580-024-00777-1] [Citation(s) in RCA: 7] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 08/23/2024] [Indexed: 01/29/2025]
Abstract
Integrin receptors are the main molecular link between cells and the extracellular matrix (ECM) as well as mediating cell-cell interactions. Integrin-ECM binding triggers the formation of heterogeneous multi-protein assemblies termed integrin adhesion complexes (IACs) that enable integrins to transform extracellular cues into intracellular signals that affect many cellular processes, especially cell motility. Cell migration is essential for diverse physiological and pathological processes and is dysregulated in cancer to favour cell invasion and metastasis. Here, we discuss recent findings on the role of integrins in cell migration with a focus on cancer cell dissemination. We review how integrins regulate the spatial distribution and dynamics of different IACs, covering classical focal adhesions, emerging adhesion types and adhesion regulation. We discuss the diverse roles integrins have during cancer progression from cell migration across varied ECM landscapes to breaching barriers such as the basement membrane, and eventual colonization of distant organs.
Collapse
Affiliation(s)
- Megan R Chastney
- Turku Bioscience Centre, University of Turku and Åbo Akademi University, Turku, Finland
| | - Jasmin Kaivola
- Turku Bioscience Centre, University of Turku and Åbo Akademi University, Turku, Finland
| | - Veli-Matti Leppänen
- Turku Bioscience Centre, University of Turku and Åbo Akademi University, Turku, Finland
| | - Johanna Ivaska
- Turku Bioscience Centre, University of Turku and Åbo Akademi University, Turku, Finland.
- Department of Life Technologies, University of Turku, Turku, Finland.
- InFLAMES Research Flagship Center, University of Turku, Turku, Finland.
- Western Finnish Cancer Center (FICAN West), University of Turku, Turku, Finland.
- Foundation for the Finnish Cancer Institute, Helsinki, Finland.
| |
Collapse
|
9
|
Roy T, Dutta S, Ghosh S, Sthanam LK, Sen S. CD44/Integrin β1 Association Drives Fast Motility on Hyaluronic Acid Substrates. J Cell Physiol 2025; 240:e70001. [PMID: 39835458 DOI: 10.1002/jcp.70001] [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: 05/24/2024] [Revised: 12/14/2024] [Accepted: 01/09/2025] [Indexed: 01/22/2025]
Abstract
In addition to proteins such as collagen (Col) and fibronectin, the extracellular matrix (ECM) is enriched with bulky proteoglycan molecules such as hyaluronic acid (HA). However, how ECM proteins and proteoglycans collectively regulate cellular processes has not been adequately explored. Here, we address this question by studying cytoskeletal and focal adhesion organization and dynamics on cells cultured on polyacrylamide hydrogels functionalized with Col, HA and a combination of Col and HA (Col/HA). We show that fastest migration on HA substrates is attributed to the presence of smaller and weaker focal adhesions. Integrinβ $\beta $ 1 co-localization and its association with CD44-which is the receptor for HA, and insensitivity of cell spreading to RGD on HA substrates suggests that focal adhesions on HA substrates are formed via integrin association with HA bound CD44. Consistent with this, adhesion formation and cell motility were inhibited when CD44 was knocked out. Collectively, our results suggest that association of integrinβ $\beta $ 1 with CD44 drives fast motility on HA substrates.
Collapse
Affiliation(s)
- Tanusri Roy
- Department of Biosciences & Bioengineering, IIT Bombay, Mumbai, India
| | - Sarbajeet Dutta
- Department of Biosciences & Bioengineering, IIT Bombay, Mumbai, India
| | - Swetlana Ghosh
- Department of Biosciences & Bioengineering, IIT Bombay, Mumbai, India
| | | | - Shamik Sen
- Department of Biosciences & Bioengineering, IIT Bombay, Mumbai, India
| |
Collapse
|
10
|
Kim C, Kang N, Min S, Thangam R, Lee S, Hong H, Kim K, Kim SY, Kim D, Rha H, Tag KR, Lee HJ, Singh N, Jeong D, Hwang J, Kim Y, Park S, Lee H, Kim T, Son SW, Park S, Karamikamkar S, Zhu Y, Hassani Najafabadi A, Chu Z, Sun W, Zhao P, Zhang K, Bian L, Song HC, Park SG, Kim JS, Lee SY, Ahn JP, Kim HK, Zhang YS, Kang H. Modularity-based mathematical modeling of ligand inter-nanocluster connectivity for unraveling reversible stem cell regulation. Nat Commun 2024; 15:10665. [PMID: 39715783 PMCID: PMC11666790 DOI: 10.1038/s41467-024-54557-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2023] [Accepted: 11/14/2024] [Indexed: 12/25/2024] Open
Abstract
The native extracellular matrix is continuously remodeled to form complex interconnected network structures that reversibly regulate stem cell behaviors. Both regulation and understanding of its intricate dynamicity can help to modulate numerous cell behaviors. However, neither of these has yet been achieved due to the lack of designing and modeling such complex structures with dynamic controllability. Here we report modularity-based mathematical modeling of extracellular matrix-emulating ligand inter-cluster connectivity using the graph theory. Increasing anisotropy of magnetic nano-blockers proportionately disconnects arginine-glycine-aspartic acid ligand-to-ligand interconnections and decreases the number of ligand inter-cluster edges. This phenomenon deactivates stem cells, which can be partly activated by linearizing the nano-blockers. Remote cyclic elevation of high-anisotropy nano-blockers flexibly generates nano-gaps under the nano-blockers and augments the number of ligand inter-cluster edges. Subsequently, integrin-presenting stem cell infiltration is stimulated, which reversibly intensifies focal adhesion and mechanotransduction-driven differentiation both in vitro and in vivo. Designing and systemically modeling extracellular matrix-mimetic geometries opens avenues for unraveling dynamic cell-material interactions for tissue regeneration.
Collapse
Affiliation(s)
- Chowon Kim
- Department of Materials Science and Engineering, Korea University, Seoul, Republic of Korea
| | - Nayeon Kang
- Department of Materials Science and Engineering, Korea University, Seoul, Republic of Korea
| | - Sunhong Min
- Department of Materials Science and Engineering, Korea University, Seoul, Republic of Korea
| | - Ramar Thangam
- Department of Materials Science and Engineering, Korea University, Seoul, Republic of Korea
| | - Sungkyu Lee
- Department of Materials Science and Engineering, Korea University, Seoul, Republic of Korea
| | - Hyunsik Hong
- Department of Materials Science and Engineering, Korea University, Seoul, Republic of Korea
| | - Kanghyeon Kim
- Department of Materials Science and Engineering, Korea University, Seoul, Republic of Korea
| | - Seong Yeol Kim
- Department of Materials Science and Engineering, Korea University, Seoul, Republic of Korea
| | - Dahee Kim
- Department of Materials Science and Engineering, Korea University, Seoul, Republic of Korea
| | - Hyunji Rha
- Department of Materials Science and Engineering, Korea University, Seoul, Republic of Korea
| | - Kyong-Ryol Tag
- Department of Materials Science and Engineering, Korea University, Seoul, Republic of Korea
- Advanced Analysis Center, Korea Institute of Science and Technology (KIST), Seoul, Republic of Korea
| | - Hyun-Jeong Lee
- Department of Materials Science and Engineering, Korea University, Seoul, Republic of Korea
- Advanced Analysis Center, Korea Institute of Science and Technology (KIST), Seoul, Republic of Korea
| | - Nem Singh
- Department of Materials Science and Engineering, Korea University, Seoul, Republic of Korea
- Department of Chemistry, Korea University, Seoul, Republic of Korea
| | - Daun Jeong
- Department of Orthopedic Surgery, Korea University Anam Hospital, Seoul, Republic of Korea
| | - Jangsun Hwang
- Department of Orthopedic Surgery, Korea University Anam Hospital, Seoul, Republic of Korea
| | - Yuri Kim
- Department of Materials Science and Engineering, Korea University, Seoul, Republic of Korea
| | - Sangwoo Park
- Department of Materials Science and Engineering, Korea University, Seoul, Republic of Korea
| | - Hyesung Lee
- Department of Chemical and Biomolecular Engineering, Yonsei University, Seoul, Republic of Korea
| | - Taeeon Kim
- Nano-Bio Convergence Department, Korea Institute of Materials Science (KIMS), Changwon, Gyeongnam, Republic of Korea
- Department of Future Convergence Materials, Korea University, Seoul, Republic of Korea
| | - Sang Wook Son
- Department of Dermatology, Korea University Ansan Hospital, Korea University College of Medicine, Ansan, Republic of Korea
| | - Steve Park
- Department of Materials Science and Engineering, Korea Advanced Institute of Science and Technology (KAIST), Daejeon, Republic of Korea
| | | | - Yangzhi Zhu
- Terasaki Institute for Biomedical Innovation, Los Angeles, CA, USA
| | | | - Zhiqin Chu
- Department of Electrical and Electronic Engineering, Joint Appointment with School of Biomedical Sciences, The University of Hong Kong, Hong Kong, PR China
| | - Wujin Sun
- Department of Biological Systems Engineering, Virginia Tech, Blacksburg, VA, USA
| | - Pengchao Zhao
- School of Biomedical Sciences and Engineering, Guangzhou International Campus, South China University of Technology, Guangzhou, PR China
| | - Kunyu Zhang
- School of Biomedical Sciences and Engineering, Guangzhou International Campus, South China University of Technology, Guangzhou, PR China
| | - Liming Bian
- School of Biomedical Sciences and Engineering, Guangzhou International Campus, South China University of Technology, Guangzhou, PR China
| | - Hyun-Cheol Song
- Electronic Materials Research Center, Korea Institute of Science and Technology (KIST), Seoul, Republic of Korea
- KIST-SKKU Carbon-Neutral Research Center, Sungkyunkwan University (SKKU), Suwon, Republic of Korea
| | - Sung-Gyu Park
- Nano-Bio Convergence Department, Korea Institute of Materials Science (KIMS), Changwon, Gyeongnam, Republic of Korea
- Department of Future Convergence Materials, Korea University, Seoul, Republic of Korea
| | - Jong Seung Kim
- Department of Chemistry, Korea University, Seoul, Republic of Korea
| | - Sang-Yup Lee
- Department of Chemical and Biomolecular Engineering, Yonsei University, Seoul, Republic of Korea
| | - Jae-Pyoung Ahn
- Advanced Analysis Center, Korea Institute of Science and Technology (KIST), Seoul, Republic of Korea
| | - Hong-Kyu Kim
- Advanced Analysis Center, Korea Institute of Science and Technology (KIST), Seoul, Republic of Korea
| | - Yu Shrike Zhang
- Division of Engineering in Medicine, Department of Medicine, Brigham and Women's Hospital Harvard Medical School, Cambridge, MA, USA.
| | - Heemin Kang
- Department of Materials Science and Engineering, Korea University, Seoul, Republic of Korea.
- Department of Future Convergence Materials, Korea University, Seoul, Republic of Korea.
- College of Medicine, Korea University, Seoul, Republic of Korea.
| |
Collapse
|
11
|
Padzińska-Pruszyńska IB, Taciak B, Kiraga Ł, Smolarska A, Górczak M, Kucharzewska P, Kubiak M, Szeliga J, Matejuk A, Król M. Targeting Cancer: Microenvironment and Immunotherapy Innovations. Int J Mol Sci 2024; 25:13569. [PMID: 39769334 PMCID: PMC11679359 DOI: 10.3390/ijms252413569] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2024] [Revised: 12/12/2024] [Accepted: 12/16/2024] [Indexed: 01/11/2025] Open
Abstract
In 2024, the United States was projected to experience 2 million new cancer diagnoses and approximately 611,720 cancer-related deaths, reflecting a broader global trend in which cancer cases are anticipated to exceed 35 million by 2050. This increasing burden highlights ongoing challenges in cancer treatment despite significant advances that have reduced cancer mortality by 31% since 1991. Key obstacles include the disease's inherent heterogeneity and complexity, such as treatment resistance, cancer stem cells, and the multifaceted tumor microenvironment (TME). The TME-comprising various tumor and immune cells, blood vessels, and biochemical factors-plays a crucial role in tumor growth and resistance to therapies. Recent innovations in cancer treatment, particularly in the field of immuno-oncology, have leveraged insights into TME interactions. An emerging example is the FDA-approved therapy using tumor-infiltrating lymphocytes (TILs), demonstrating the potential of cell-based approaches in solid tumors. However, TIL therapy is just one of many strategies being explored. This review provides a comprehensive overview of the emerging field of immuno-oncology, focusing on how novel therapies targeting or harnessing components of the TME could enhance treatment efficacy and address persistent challenges in cancer care.
Collapse
Affiliation(s)
- Irena Barbara Padzińska-Pruszyńska
- Center of Cellular Immunotherapies, Warsaw University of Life Sciences, 02-787 Warsaw, Poland; (I.B.P.-P.); (B.T.); (A.S.); (M.G.); (P.K.); (M.K.); (J.S.)
| | - Bartłomiej Taciak
- Center of Cellular Immunotherapies, Warsaw University of Life Sciences, 02-787 Warsaw, Poland; (I.B.P.-P.); (B.T.); (A.S.); (M.G.); (P.K.); (M.K.); (J.S.)
| | - Łukasz Kiraga
- Division of Pharmacology and Toxicology, Department of Preclinical Sciences, Institute of Veterinary Medicine, Warsaw University of Life Sciences, 02-787 Warsaw, Poland;
| | - Anna Smolarska
- Center of Cellular Immunotherapies, Warsaw University of Life Sciences, 02-787 Warsaw, Poland; (I.B.P.-P.); (B.T.); (A.S.); (M.G.); (P.K.); (M.K.); (J.S.)
| | - Małgorzata Górczak
- Center of Cellular Immunotherapies, Warsaw University of Life Sciences, 02-787 Warsaw, Poland; (I.B.P.-P.); (B.T.); (A.S.); (M.G.); (P.K.); (M.K.); (J.S.)
| | - Paulina Kucharzewska
- Center of Cellular Immunotherapies, Warsaw University of Life Sciences, 02-787 Warsaw, Poland; (I.B.P.-P.); (B.T.); (A.S.); (M.G.); (P.K.); (M.K.); (J.S.)
| | - Małgorzata Kubiak
- Center of Cellular Immunotherapies, Warsaw University of Life Sciences, 02-787 Warsaw, Poland; (I.B.P.-P.); (B.T.); (A.S.); (M.G.); (P.K.); (M.K.); (J.S.)
| | - Jacek Szeliga
- Center of Cellular Immunotherapies, Warsaw University of Life Sciences, 02-787 Warsaw, Poland; (I.B.P.-P.); (B.T.); (A.S.); (M.G.); (P.K.); (M.K.); (J.S.)
| | - Agata Matejuk
- Department of Immunology, Collegium Medicum, University of Zielona Góra, 65-046 Zielona Góra, Poland;
| | - Magdalena Król
- Center of Cellular Immunotherapies, Warsaw University of Life Sciences, 02-787 Warsaw, Poland; (I.B.P.-P.); (B.T.); (A.S.); (M.G.); (P.K.); (M.K.); (J.S.)
| |
Collapse
|
12
|
Bach-Griera M, Hernández A, Julián E. Mycobacteria Treatment Inhibits Bladder Cancer Cell Migration, Invasion, and Anchorage-Independent Growth. Int J Mol Sci 2024; 25:12997. [PMID: 39684712 DOI: 10.3390/ijms252312997] [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: 09/23/2024] [Revised: 11/26/2024] [Accepted: 11/28/2024] [Indexed: 12/18/2024] Open
Abstract
Bladder cancer (BC) is a highly recurrent and invasive malignancy, with Mycobacterium bovis BCG serving as the primary immunotherapy, particularly for non-muscle-invasive bladder cancer (NMIBC). However, the mechanisms underlying BCG's antitumor effects and the potential of non-tuberculous mycobacteria like Mycobacterium brumae remain unclear. This study investigates the antitumor effects of M. bovis BCG and M. brumae on BC cell migration, invasion, and anchorage-independent growth. BC cell lines representing different stages of tumor differentiation were treated with either M. bovis BCG or M. brumae. Cell migration was assessed through wound healing and transwell assays, invasiveness by transwell invasion assays, MMP-9 production by gelatin zymography, and anchorage-independent growth via soft agar colony formation. Both mycobacteria inhibited individual cell migration across all BC lines, while collective migration was only reduced in intermediate-grade cells. Both treatments also reduced invasiveness, associated with decreased MMP-9 production. Furthermore, M. brumae inhibited anchorage-independent growth across all BC lines, while M. bovis BCG had a more selective effect, primarily inhibiting growth in high-grade cells. In conclusion, both mycobacteria reduce migration, invasion, and anchorage-independent growth of BC cells, with their effectiveness varying by species and tumor differentiation grade.
Collapse
Affiliation(s)
- Marc Bach-Griera
- Microbiology Unit, Department of Genetics and Microbiology, Biosciences School, Universitat Autonoma de Barcelona, 08193 Bellaterra, Spain
| | - Alba Hernández
- Genetics Unit, Department of Genetics and Microbiology, Biosciences School, Universitat Autonoma de Barcelona, 08193 Bellaterra, Spain
| | - Esther Julián
- Microbiology Unit, Department of Genetics and Microbiology, Biosciences School, Universitat Autonoma de Barcelona, 08193 Bellaterra, Spain
| |
Collapse
|
13
|
Chang CW, Bale A, Bhargava R, Harley BAC. Glioblastoma Drives Protease-Independent Extracellular Matrix Invasion of Microglia. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.11.08.622715. [PMID: 39605524 PMCID: PMC11601220 DOI: 10.1101/2024.11.08.622715] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/29/2024]
Abstract
Glioblastoma (GBM) is the most common and lethal form of primary brain cancer. Microglia infiltration into the tumor microenvironment is associated with immunosuppression and poor prognosis. Improved physicochemical understanding of microglia activation and invasion may provide novel GBM therapeutic strategies essential for improving long-term treatment efficacy. Here, we combine microfluidic systems with 3-D collagen hydrogels to systematically investigate microglia activation, invasion, contractility and cytokine secretion in response of GBM-microglia crosstalk. GBM inflammatory biomolecules significantly promote activation and 3D invasion of microglia. Interestingly, microglia invasion is not significantly affected by inhibitors of MMP activity or cellular glycolysis. In contrast, ROCK-pathway inhibition significantly impedes microglia invasion. Infrared microscopy analyses show that GBM co-culture does not significantly alter microglia lipid content. Further, GBM conditioned media resulted in significantly increased collagen hydrogel contraction, suggesting the importance of microglia contractility to physically remodel the local extracellular matrix (ECM). We also identify a panel of soluble proteins that may contribute to microglia chemotaxis, such as TIMP-1 and CXCL12. Taken together, this study suggests that the presence of GBM cells can enhance microglia invasion via increased cellular contractility, independent of MMP activity and cellular glycolysis.
Collapse
|
14
|
Sapudom J, Riedl P, Schricker M, Kroy K, Pompe T. Physical network regimes of 3D fibrillar collagen networks trigger invasive phenotypes of breast cancer cells. BIOMATERIALS ADVANCES 2024; 163:213961. [PMID: 39032434 DOI: 10.1016/j.bioadv.2024.213961] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/22/2024] [Revised: 06/18/2024] [Accepted: 07/14/2024] [Indexed: 07/23/2024]
Abstract
The mechanical characteristics of the extracellular environment are known to significantly influence cancer cell behavior in vivo and in vitro. The structural complexity and viscoelastic dynamics of the extracellular matrix (ECM) pose significant challenges in understanding its impact on cancer cells. Herein, we report distinct regulatory signatures in the invasion of different breast cancer cell lines into three-dimensional (3D) fibrillar collagen networks, caused by systematic modifications of the physical network properties. By reconstituting collagen networks of thin fibrils, we demonstrate that such networks can display network strand flexibility akin to that of synthetic polymer networks, known to exhibit entropic rubber elasticity. This finding contrasts with the predominant description of the mechanics of fibrillar collagen networks by an enthalpic bending elasticity of rod-like fibrils. Mean-squared displacement analysis of free-standing fibrils confirmed a flexible fiber regime in networks of thin fibrils. Furthermore, collagen fibrils in both networks were softened by the adsorption of highly negatively charged sulfonated polymers and colloidal probe force measurements of network elastic modulus again proofed the occurrence of the two different physical network regimes. Our cell assays revealed that the cellular behavior (morphology, clustering, invasiveness, matrix metalloproteinase (MMP) activity) of the 'weakly invasive' MCF-7 and 'highly invasive' MDA-MB-231 breast cancer cell lines is distinctively affected by the physical (enthalpic/entropic) network regime, and cannot be explained by changes of the network elastic modulus, alone. These results highlight an essential pathway, albeit frequently overlooked, how the physical characteristics of fibrillar ECMs affect cellular behavior. Considering the coexistence of diverse physical network regimes of the ECM in vivo, our findings underscore their critical role of ECM's physical network regimes in tumor progression and other cell functions, and moreover emphasize the significance of 3D in vitro collagen network models for quantifying cell responses in both healthy and pathological states.
Collapse
Affiliation(s)
- Jiranuwat Sapudom
- Institute of Biochemistry, Leipzig University, 04103 Leipzig, Germany; Division of Engineering, New York University Abu Dhabi, Abu Dhabi, United Arab Emirates
| | - Philipp Riedl
- Institute of Biochemistry, Leipzig University, 04103 Leipzig, Germany
| | - Maria Schricker
- Institute of Biochemistry, Leipzig University, 04103 Leipzig, Germany
| | - Klaus Kroy
- Institute for Theoretical Physics, Leipzig University, Leipzig 04009, Germany
| | - Tilo Pompe
- Institute of Biochemistry, Leipzig University, 04103 Leipzig, Germany.
| |
Collapse
|
15
|
Arpinati L, Carradori G, Scherz-Shouval R. CAF-induced physical constraints controlling T cell state and localization in solid tumours. Nat Rev Cancer 2024; 24:676-693. [PMID: 39251836 DOI: 10.1038/s41568-024-00740-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 08/05/2024] [Indexed: 09/11/2024]
Abstract
Solid tumours comprise cancer cells that engage in continuous interactions with non-malignant cells and with acellular components, forming the tumour microenvironment (TME). The TME has crucial and diverse roles in tumour progression and metastasis, and substantial efforts have been dedicated into understanding the functions of different cell types within the TME. These efforts highlighted the importance of non-cell-autonomous signalling in cancer, mediating interactions between the cancer cells, the immune microenvironment and the non-immune stroma. Much of this non-cell-autonomous signalling is mediated through acellular components of the TME, known as the extracellular matrix (ECM), and controlled by the cells that secrete and remodel the ECM - the cancer-associated fibroblasts (CAFs). In this Review, we delve into the complex crosstalk among cancer cells, CAFs and immune cells, highlighting the effects of CAF-induced ECM remodelling on T cell functions and offering insights into the potential of targeting ECM components to improve cancer therapies.
Collapse
Affiliation(s)
- Ludovica Arpinati
- Department of Biomolecular Sciences, The Weizmann Institute of Science, Rehovot, Israel
| | - Giulia Carradori
- Department of Biomolecular Sciences, The Weizmann Institute of Science, Rehovot, Israel
| | - Ruth Scherz-Shouval
- Department of Biomolecular Sciences, The Weizmann Institute of Science, Rehovot, Israel.
| |
Collapse
|
16
|
Lehmann M, Weixler B, Elezkurtaj S, Loddenkemper C, Kühl AA, Siegmund B. Spatial Single Cell Profiling Using Imaging Mass Cytometry: Inflammatory Versus Penetrating Crohn's Disease. J Crohns Colitis 2024; 18:1305-1318. [PMID: 38465390 DOI: 10.1093/ecco-jcc/jjae033] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/05/2024] [Revised: 02/14/2024] [Accepted: 03/07/2024] [Indexed: 03/12/2024]
Abstract
BACKGROUND AND AIMS Fistula formation is a major complication in Crohn's disease [CD] and the role of the immune cell compartment remains to be elucidated. Thus, we compared the immune cell compartment of CD fistula to inflammatory CD colitis using imaging mass cytometry [IMC] and immunofluorescence. METHODS A 36-marker panel including structural, functional, and lineage markers for use in IMC was established. This panel was applied to analyse paraffin-embedded CD fistula tract [n = 11], CD colitis [n = 10], and colon samples from non-inflamed controls [n = 12]. Computational methods for cell segmentation, dimensionality reduction, and cell type clustering were used to define cell populations for cell frequency, marker distribution, and spatial neighbourhood analysis. Multiplex immunofluorescence was used for higher resolution spatial analysis. RESULTS Analysis of cell frequencies in CD fistulas compared to CD colitis and control colonic samples revealed a significant increase in neutrophils, effector cytotoxic T cells, and inflammatory macrophages in CD fistula samples, whereas regulatory T cells were decreased. Neutrophils in CD fistula expressed significantly more matrix metalloproteinase 9 [MMP9], correlating with extracellular matrix remodelling. Neighbourhood analysis revealed a strong association between MMP9+ neutrophils and effector cytotoxic T cells in both CD fistulas and colitis. CONCLUSIONS This study presents the first highly multiplexed single cell analysis of the immune cell compartment of CD fistulas and their spatial context. It links immune cell dynamics, particularly MMP9+ neutrophils, to extracellular matrix remodelling in CD fistulas, offering insights into the complex network of cellular interactions and potential therapeutic targets for CD complications.
Collapse
Affiliation(s)
- Malte Lehmann
- Department of Gastroenterology, Infectious Diseases and Rheumatology, Charité - Universitätsmedizin Berlin, corporate member of Freie Universität Berlin, Humboldt-Universität zu Berlin, Campus Benjamin Franklin, Hindenburgdamm 30, 12200, Berlin, Germany
| | - Benjamin Weixler
- Department of General and Visceral Surgery, Charité - Universitätsmedizin Berlin, corporate member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Berlin, Germany
| | - Sefer Elezkurtaj
- Institute of Pathology, Charité - Universitätsmedizin Berlin, corporate member of Freie Universität Berlin, Humboldt-Universität zu Berlin, Berlin, Germany
| | - Christopher Loddenkemper
- PathoTres, Gemeinschaftspraxis für Pathologie und Neuropathologie, Teltowkanalstr. 2, 12247, Berlin, Germany
| | - Anja A Kühl
- iPATH.Berlin, Campus Benjamin Franklin, Charité - Universitätsmedizin Berlin, corporate member of Freie Universität Berlin, Humboldt-Universität zu Berlin, Berlin, Germany
| | - Britta Siegmund
- Department of Gastroenterology, Infectious Diseases and Rheumatology, Charité - Universitätsmedizin Berlin, corporate member of Freie Universität Berlin, Humboldt-Universität zu Berlin, Campus Benjamin Franklin, Hindenburgdamm 30, 12200, Berlin, Germany
| |
Collapse
|
17
|
Jin Y, Christenson ES, Zheng L, Li K. Neutrophils in pancreatic ductal adenocarcinoma: bridging preclinical insights to clinical prospects for improved therapeutic strategies. Expert Rev Clin Immunol 2024; 20:945-958. [PMID: 38690749 DOI: 10.1080/1744666x.2024.2348605] [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: 01/20/2024] [Accepted: 04/24/2024] [Indexed: 05/03/2024]
Abstract
INTRODUCTION Pancreatic ductal adenocarcinoma (PDAC) is a highly aggressive malignancy characterized by a dismal five-year survival rate of less than 10%. Neutrophils are key components of the innate immune system, playing a pivotal role in the PDAC immune microenvironment. AREAS COVERED This review provides a comprehensive survey of the pivotal involvement of neutrophils in the tumorigenesis and progression of PDAC. Furthermore, it synthesizes preclinical and clinical explorations aimed at targeting neutrophils within the milieu of PDAC, subsequently proposing a conceptual framework to propel further inquiry focused on enhancing the therapeutic efficacy of PDAC through neutrophil-targeted strategies. PubMed and Web of Science databases were utilized for researching neutrophils in pancreatic cancer publications prior to 2024. EXPERT OPINION Neutrophils play roles in promoting tumor growth and metastasis in PDAC and are associated with poor prognosis. However, the heterogeneity and plasticity of neutrophils and their complex relationships with other immune cells and extracellular matrix also provide new insights for immunotherapy targeting neutrophils to achieve a better prognosis for PDAC.
Collapse
Affiliation(s)
- Yi Jin
- Division of Pancreatic Surgery, Department of General Surgery, West China Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Eric S Christenson
- The Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University School of Medicine, Baltimore, MD, USA
- The Pancreatic Cancer Precision Medicine Center of Excellence Program, Johns Hopkins University School of Medicine, Baltimore, MD, USA
- The Bloomberg Kimmel Institute for Cancer Immunotherapy, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Lei Zheng
- The Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University School of Medicine, Baltimore, MD, USA
- The Pancreatic Cancer Precision Medicine Center of Excellence Program, Johns Hopkins University School of Medicine, Baltimore, MD, USA
- The Bloomberg Kimmel Institute for Cancer Immunotherapy, Johns Hopkins University School of Medicine, Baltimore, MD, USA
- Department of Oncology, Johns Hopkins University School of Medicine, Baltimore, MD, USA
- The Skip Viragh Pancreatic Cancer Center, Johns Hopkins University School of Medicine, Baltimore, MD, USA
- The Cancer Convergence Institute at Johns Hopkins, Johns Hopkins University School of Medicine, Baltimore, MD, USA
- Department of Surgery, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Keyu Li
- Division of Pancreatic Surgery, Department of General Surgery, West China Hospital, Sichuan University, Chengdu, Sichuan, China
- The Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University School of Medicine, Baltimore, MD, USA
- The Pancreatic Cancer Precision Medicine Center of Excellence Program, Johns Hopkins University School of Medicine, Baltimore, MD, USA
- The Bloomberg Kimmel Institute for Cancer Immunotherapy, Johns Hopkins University School of Medicine, Baltimore, MD, USA
- The Skip Viragh Pancreatic Cancer Center, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| |
Collapse
|
18
|
Narasimhan BN, Fraley SI. Degradability tunes ECM stress relaxation and cellular mechanics. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.07.28.605514. [PMID: 39131364 PMCID: PMC11312499 DOI: 10.1101/2024.07.28.605514] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 08/13/2024]
Abstract
In native extracellular matrices (ECM), cells can use matrix metalloproteinases (MMPs) to degrade and remodel their surroundings. Likewise, synthetic matrices have been engineered to facilitate MMP-mediated cleavage that enables cell spreading, migration, and interactions. However, the intersection of matrix degradability and mechanical properties has not been fully considered. We hypothesized that immediate mechanical changes result from the action of MMPs on the ECM and that these changes are sensed by cells. Using atomic force microscopy (AFM) to measure cell-scale mechanical properties, we find that both fibrillar collagen and synthetic degradable matrices exhibit enhanced stress relaxation after MMP exposure. Cells respond to these relaxation differences by altering their spreading and focal adhesions. We demonstrate that stress relaxation can be tuned through the rational design of matrix degradability. These findings establish a fundamental link between matrix degradability and stress relaxation, which may impact a range of biological applications.
Collapse
Affiliation(s)
| | - Stephanie I. Fraley
- Department of Bioengineering, University of California, San Diego, La Jolla, CA 92093, USA
| |
Collapse
|
19
|
Kong J, Deng Y, Xu Y, Zhang P, Li L, Huang Y. A Two-Pronged Delivery Strategy Disrupting Positive Feedback Loop of Neutrophil Extracellular Traps for Metastasis Suppression. ACS NANO 2024; 18:15432-15451. [PMID: 38842256 DOI: 10.1021/acsnano.3c09165] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/07/2024]
Abstract
Neutrophil extracellular traps (NETs) severely affect tumor metastasis through a self-perpetuating feedback loop involving two key steps: (1) mitochondrial aerobic respiration-induced hypoxia promotes NET formation and (2) NETs enhance mitochondrial metabolism to exacerbate hypoxia. Herein, we propose a two-pronged approach with the activity of NET-degrading and mitochondrion-damaging by simultaneously targeting drugs to NETs and tumor mitochondria of this loop. In addition to specifically recognizing and eliminating extant NETs, the NET-targeting nanoparticle also reduces NET-induced mitochondrial biogenesis, thus inhibiting the initial step of the feedback loop and mitigating extant NETs' impact on tumor metastasis. Simultaneously, the mitochondrion-targeting system intercepts mitochondrial metabolism and alleviates tumor hypoxia, inhibiting neutrophil infiltration and subsequent NET formation, which reduces the source of NETs and disrupts another step of the self-amplifying feedback loop. Together, the combination significantly reduces the formation of NET-tumor cell clusters by disrupting the interaction between NETs and tumor mitochondria, thereby impeding the metastatic cascade including tumor invasion, hematogenous spread, and distant colonization. This work represents an innovative attempt to disrupt the feedback loop in tumor metastasis, offering a promising therapeutic approach restraining NET-assisted metastasis.
Collapse
Affiliation(s)
- Jinxia Kong
- Key Laboratory of Drug-Targeting and Drug Delivery System of the Education Ministry and Sichuan Province, Sichuan Engineering Laboratory for Plant-Sourced Drug and Sichuan Research Center for Drug Precision Industrial Technology, West China School of Pharmacy, Sichuan University, Chengdu 610041, China
| | - Yudi Deng
- Key Laboratory of Drug-Targeting and Drug Delivery System of the Education Ministry and Sichuan Province, Sichuan Engineering Laboratory for Plant-Sourced Drug and Sichuan Research Center for Drug Precision Industrial Technology, West China School of Pharmacy, Sichuan University, Chengdu 610041, China
| | - Yiwen Xu
- Key Laboratory of Drug-Targeting and Drug Delivery System of the Education Ministry and Sichuan Province, Sichuan Engineering Laboratory for Plant-Sourced Drug and Sichuan Research Center for Drug Precision Industrial Technology, West China School of Pharmacy, Sichuan University, Chengdu 610041, China
| | - Ping Zhang
- Key Laboratory of Drug-Targeting and Drug Delivery System of the Education Ministry and Sichuan Province, Sichuan Engineering Laboratory for Plant-Sourced Drug and Sichuan Research Center for Drug Precision Industrial Technology, West China School of Pharmacy, Sichuan University, Chengdu 610041, China
| | - Lian Li
- Key Laboratory of Drug-Targeting and Drug Delivery System of the Education Ministry and Sichuan Province, Sichuan Engineering Laboratory for Plant-Sourced Drug and Sichuan Research Center for Drug Precision Industrial Technology, West China School of Pharmacy, Sichuan University, Chengdu 610041, China
| | - Yuan Huang
- Key Laboratory of Drug-Targeting and Drug Delivery System of the Education Ministry and Sichuan Province, Sichuan Engineering Laboratory for Plant-Sourced Drug and Sichuan Research Center for Drug Precision Industrial Technology, West China School of Pharmacy, Sichuan University, Chengdu 610041, China
| |
Collapse
|
20
|
Zhu W, Zhao S, Cheng X, Wu C, Liu Z, Huang J. Chemokine‑ and chemokine receptor-based subtypes predict prognosis, immunotherapy and chemotherapy response in colorectal cancer patients. Int Immunopharmacol 2024; 134:112172. [PMID: 38703566 DOI: 10.1016/j.intimp.2024.112172] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2024] [Revised: 04/12/2024] [Accepted: 04/27/2024] [Indexed: 05/06/2024]
Abstract
BACKGROUND The clinical significance and comprehensive characteristics of chemokines and chemokine receptors in colorectal cancer (CRC) have not been previously reported. Our study aims to investigate the expression profiles of chemokines and chemokine receptors, as well as establish subtypes in CRC. METHODS 1009 CRC samples were enrolled in our study. Consensus unsupervised clustering analysis was conducted to establish subtypes, and a risk score model was developed using univariate Cox regression and least absolute shrinkage and selection operator (LASSO) analyses. 36 pairs of tissue specimens of CRC patients and two CRC cell lines were used to validate the subtypes and risk score in vitro. Quantitative real-time PCR and western blotting were employed to validate mRNA and protein expression levels, respectively. Flow cytometry was utilized for analyzing cell apoptosis, while cell viability assay and EdU assay were conducted to assess cell proliferation ability. RESULTS The Cluster B group shares similarities with the low-risk group in terms of exhibiting a higher level of immune cell infiltration and belonging to hot tumor. Patients CRC in the Cluster B group demonstrate a more favorable prognosis and exhibit better response to immunotherapy and chemotherapy. On the other hand, the Cluster A group resembles the high-risk group as it displays lower levels of immune cell infiltration, indicating a cold tumor phenotype. CRC patients in the Cluster A group have poorer prognoses and show less therapeutic efficacy towards immunotherapy and chemotherapy. Furthermore, we utilized a total of 36 pairs of tissue samples obtained from patients with CRC, along with two CRC cell lines for validation in vitro. This comprehensive approach further enhances the scientific validity and reliability of the identified subtypes and risk score in their ability to predict prognosis, response to immunotherapy, and response to chemotherapy among CRC patients. CONCLUSION We first established robust prognostic subtypes based on chemokines and chemokine receptors, which could potentially serve as a novel biomarker for guiding individualized treatment in patients with CRC undergoing immunotherapy and chemotherapy.
Collapse
Affiliation(s)
- Wenjie Zhu
- Department of Gastrointestinal Surgery, The Second Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, Jiangxi Province, China; Jiangxi Province Key Laboratory of Molecular Medicine, The Second Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, Jiangxi Province, China
| | - Shimin Zhao
- Department of Gastrointestinal Surgery, The Second Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, Jiangxi Province, China; Jiangxi Province Key Laboratory of Molecular Medicine, The Second Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, Jiangxi Province, China
| | - Xiufeng Cheng
- Department of Critical Care Medicine, The First Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, Jiangxi Province, China
| | - Changlei Wu
- Department of Gastrointestinal Surgery, The Second Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, Jiangxi Province, China; Jiangxi Province Key Laboratory of Molecular Medicine, The Second Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, Jiangxi Province, China
| | - Zitao Liu
- Department of Gastrointestinal Surgery, The Second Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, Jiangxi Province, China; Jiangxi Province Key Laboratory of Molecular Medicine, The Second Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, Jiangxi Province, China
| | - Jun Huang
- Department of Gastrointestinal Surgery, The Second Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, Jiangxi Province, China.
| |
Collapse
|
21
|
Yu Z, Xiong Z, Ma J, Du P, Wang S, Liu J, Cao Y, Yang Y. Prognostic and clinicopathological significance of systemic immune-inflammation index in upper tract urothelial carcinoma: a meta-analysis of 3911 patients. Front Oncol 2024; 14:1342996. [PMID: 38947894 PMCID: PMC11211359 DOI: 10.3389/fonc.2024.1342996] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2023] [Accepted: 05/24/2024] [Indexed: 07/02/2024] Open
Abstract
Background Systemic immune-inflammation index (SII), a novel prognostic indicator, is being more commonly utilized in different types of cancer. This research project involved combining information from previously published studies to examine how pre-treatment SII can predict outcomes in individuals with upper tract urothelial carcinoma (UTUC). Further examination of the correlation between SII and clinical and pathological features in UTUC. Methods We thoroughly chose pertinent articles from various databases including PubMed, Embase, Cochrane Library, Web of Science, Chinese National Knowledge Infrastructure (CNKI), WanFang database, and Chinese Scientific Journal Database (VIP) until March 10, 2022.The data collected was analyzed using Stata 17.0 software (Stat Corp, College Station, TX). Subsequently, the impact of SII on the survival outcomes of UTUC patients was evaluated by combining HRs with 95% confidence intervals. Results Six included studies were finally confirmed, including 3911 UTUC patients in seven cohorts. The results showed that high SII before treatment predicted poor overall survival (HR =1.87, 95%CI 1.20-2.92, p=0.005), cancer specific survival (HR=2.70, 95%CI 1.47-4.96, P=0.001), and recurrence-free survival (HR =1.52, 95%CI 1.12-2.07, P=0.007). And the elevated SII may be related to LVI (present vs. absent) (OR=0.83, 95% CI=0.71-0.97, p=0.018), pT stage (pT ≥3 vs. < 3) (OR=1.82, 95% CI=1.21-2.72, p=0.004), and pN stage (N+ vs. N0) (OR=3.27, 95% CI=1.60-6.71, p=0.001). Conclusion A comprehensive analysis of all included articles in this study showed that higher pretreatment SII was related to poorer survival outcomes and adverse pathological features independently. Systematic review registration https://www.crd.york.ac.uk/prospero/, identifier CRD42022316333.
Collapse
Affiliation(s)
- Ziyi Yu
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education), Urological Department, Peking University Cancer Hospital and Institute, Beijing, China
| | - Zhencheng Xiong
- Trauma Medical Center, Department of Orthopedics Surgery, West China Hospital, West China Medical School, Sichuan University, Chengdu, China
| | - Jinchao Ma
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education), Urological Department, Peking University Cancer Hospital and Institute, Beijing, China
| | - Peng Du
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education), Urological Department, Peking University Cancer Hospital and Institute, Beijing, China
| | - Shuo Wang
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education), Urological Department, Peking University Cancer Hospital and Institute, Beijing, China
| | - Jia Liu
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education), Urological Department, Peking University Cancer Hospital and Institute, Beijing, China
| | - Yudong Cao
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education), Urological Department, Peking University Cancer Hospital and Institute, Beijing, China
| | - Yong Yang
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education), Urological Department, Peking University Cancer Hospital and Institute, Beijing, China
| |
Collapse
|
22
|
Issa H, Loubaki L, Al Amri A, Zibara K, Almutairi MH, Rouabhia M, Semlali A. Eugenol as a potential adjuvant therapy for gingival squamous cell carcinoma. Sci Rep 2024; 14:10958. [PMID: 38740853 DOI: 10.1038/s41598-024-60754-8] [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: 09/27/2023] [Accepted: 04/26/2024] [Indexed: 05/16/2024] Open
Abstract
Adoption of plant-derived compounds for the management of oral cancer is encouraged by the scientific community due to emerging chemoresistance and conventional treatments adverse effects. Considering that very few studies investigated eugenol clinical relevance for gingival carcinoma, we ought to explore its selectivity and performance according to aggressiveness level. For this purpose, non-oncogenic human oral epithelial cells (GMSM-K) were used together with the Tongue (SCC-9) and Gingival (Ca9-22) squamous cell carcinoma lines to assess key tumorigenesis processes. Overall, eugenol inhibited cell proliferation and colony formation while inducing cytotoxicity in cancer cells as compared to normal counterparts. The recorded effect was greater in gingival carcinoma and appears to be mediated through apoptosis induction and promotion of p21/p27/cyclin D1 modulation and subsequent Ca9-22 cell cycle arrest at the G0/G1 phase, in a p53-independent manner. At these levels, distinct genetic profiles were uncovered for both cell lines by QPCR array. Moreover, it seems that our active component limited Ca9-22 and SCC-9 cell migration respectively through MMP1/3 downregulation and stimulation of inactive MMPs complex formation. Finally, Ca9-22 behaviour appears to be mainly modulated by the P38/STAT5/NFkB pathways. In summary, we can disclose that eugenol is cancer selective and that its mediated anti-cancer mechanisms vary according to the cell line with gingival squamous cell carcinoma being more sensitive to this phytotherapy agent.
Collapse
Affiliation(s)
- Hawraa Issa
- GREB Research Group, Faculty of Dentistry, Laval University, Québec, Canada
| | - Lionel Loubaki
- Héma-Québec, Medical Affairs and Innovation, Québec, Canada
| | - Abdullah Al Amri
- Biochemistry Department, College of Science, King Saud University, Riyadh, Saudi Arabia
| | - Kazem Zibara
- PRASE and Biology Department, Faculty of Sciences-I, Lebanese University, Beirut, Lebanon
| | - Mikhlid H Almutairi
- Zoology Department, College of Science, King Saud University, Riyadh, Saudi Arabia
| | - Mahmoud Rouabhia
- GREB Research Group, Faculty of Dentistry, Laval University, Québec, Canada
| | - Abdelhabib Semlali
- GREB Research Group, Faculty of Dentistry, Laval University, Québec, Canada.
| |
Collapse
|
23
|
Shi S, Wang Y, Wu J, Zha B, Li P, Liu Y, Yang Y, Kong J, Gao S, Cui H, Huangfu L, Sun X, Li Z, Liang T, Zheng Y, Yang D. Predictive value of PD-L1 and TMB for short-term efficacy prognosis in non-small cell lung cancer and construction of prediction models. Front Oncol 2024; 14:1342262. [PMID: 38756661 PMCID: PMC11096522 DOI: 10.3389/fonc.2024.1342262] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2023] [Accepted: 04/08/2024] [Indexed: 05/18/2024] Open
Abstract
Objective To investigate the correlation between programmed death ligand 1(PD-L1), tumor mutation burden (TMB) and the short-term efficacy and clinical characteristics of anti-PD-1 immune checkpoint inhibitor combination chemotherapy in NSCLC patients. The efficacy of the prediction model was evaluated. Methods A total of 220 NSCLC patients receiving first-line treatment with anti-PD-1 immune checkpoint inhibitor combined with chemotherapy were retrospectively collected. The primary endpoint was short-term efficacy ORR. The correlation between short-term efficacy, PD-L1, TMB, and clinical characteristics using χ2 test or t-test was evaluated. Screen the independent prognostic factors using univariate and multivariate logistic regression analyses, and construct a nomogram prediction model using the "rms" package in R software. Using receiver operating characteristic (ROC) curve analysis to evaluate the independent Prognostic factors and the prediction model. Using decision curve analysis (DCA) to verify the superiority of the prediction model. Results The mean values of PD-L1, TMB, neutrophils, lymphocytes, neutrophil-to-lymphocyte ratio, and albumin were the highest in the ORR group, PD-L1 expression and TMB correlated with epidermal growth factor receptor expression. Multivariate analyses showed that PD-L1, TMB, and neutrophil were independent prognostic factors for ORR. The area under the ROC curve (AUC) values of the ROC constructed based on these three indicators were 0.7104, 0.7139, and 0.7131, respectively. The AUC value under the ROC of the nomogram model was 0.813. The DCA of the model showed that all three indicators used together to build the prediction model of the net return were higher than those of the single indicator prediction model. Conclusion PD-L1, TMB, and neutrophils are independent prognostic factors for short-term efficacy. The nomogram prediction model constructed using these three indicators can further improve predictive efficacy of ICIs in patients with NSCLC.
Collapse
Affiliation(s)
- Shuling Shi
- Department of Radiation Oncology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China
| | - Yingyi Wang
- Department of Radiation Oncology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China
| | - Jingjing Wu
- Department of Radiation Oncology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China
| | - Boya Zha
- Department of Radiation Oncology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China
| | - Peihong Li
- Department of Radiation Oncology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China
| | - Yukun Liu
- Department of Radiation Oncology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China
| | - Yuchuan Yang
- Department of Radiation Oncology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China
| | - Jinglin Kong
- Department of Radiation Oncology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China
| | - Shibo Gao
- Department of Radiation Oncology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China
| | - Haiyang Cui
- Department of Radiation Oncology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China
| | - Linkuan Huangfu
- Department of Radiation Oncology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China
| | - Xiaocong Sun
- Department of Radiation Oncology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China
| | - Zhikai Li
- Department of Radiation Oncology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China
| | - Tiansong Liang
- Department of Radiation Oncology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China
| | - Yingjuan Zheng
- Department of Radiation Oncology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China
- Institute of Radiotherapy and Critical Care Oncology, Zhengzhou University, Zhengzhou, Henan, China
| | - Daoke Yang
- Department of Radiation Oncology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China
- Institute of Radiotherapy and Critical Care Oncology, Zhengzhou University, Zhengzhou, Henan, China
| |
Collapse
|
24
|
Gayan S, Teli A, Sonawane A, Dey T. Impact of Chemotherapeutic Stress Depends on The Nature of Breast Cancer Spheroid and Induce Behavioral Plasticity to Resistant Population. Adv Biol (Weinh) 2024; 8:e2300271. [PMID: 38063815 DOI: 10.1002/adbi.202300271] [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: 07/01/2023] [Revised: 11/20/2023] [Indexed: 04/15/2024]
Abstract
Cellular or tumor dormancy, identified recently as one of the main reasons behind post-therapy recurrence, can be caused by diverse reasons. Chemotherapy has recently been recognized as one of such reasons. However, in-depth studies of chemotherapy-induced dormancy are lacking due to the absence of an in vitro human-relevant model tailor-made for such a scenario. This report utilized multicellular breast cancer spheroid to create a primary platform for establishing a chemotherapy-induced dormancy model. It is observed that extreme chemotherapeutic stress affects invasive and non-invasive spheroids differently. Non-invasive spheroids exhibit more resilience and maintain viability and migrational ability, while invasive spheroids display heightened susceptibility and improved tumorigenic capacity. Heterogenous spheroids exhibit increased tumorigenic capacity while show minimal survival ability. Further probing of chemotherapeutically dormant spheroids is needed to understand the molecular mechanism and identify dormancy-related markers to achieve therapeutic success in the future.
Collapse
Affiliation(s)
- Sukanya Gayan
- Department of Biotechnology (merged with Institute of Bioinformatics and Biotechnology), Savitribai Phule Pune University, Pune, 411007, India
| | - Abhishek Teli
- Department of Biotechnology (merged with Institute of Bioinformatics and Biotechnology), Savitribai Phule Pune University, Pune, 411007, India
| | - Akshay Sonawane
- Department of Biotechnology (merged with Institute of Bioinformatics and Biotechnology), Savitribai Phule Pune University, Pune, 411007, India
| | - Tuli Dey
- Department of Biotechnology (merged with Institute of Bioinformatics and Biotechnology), Savitribai Phule Pune University, Pune, 411007, India
| |
Collapse
|
25
|
Yang S, Jia J, Wang F, Wang Y, Fang Y, Yang Y, Zhou Q, Yuan W, Bian Z. Targeting neutrophils: Mechanism and advances in cancer therapy. Clin Transl Med 2024; 14:e1599. [PMID: 38450975 PMCID: PMC10918741 DOI: 10.1002/ctm2.1599] [Citation(s) in RCA: 10] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2023] [Revised: 02/03/2024] [Accepted: 02/07/2024] [Indexed: 03/08/2024] Open
Abstract
BACKGROUND Cancer is a thorny problem which cannot be conquered by mankind at present and recent researchers have put their focus on tumor microenviroment. Neutrophils, the prominent leukocytes in peripheral blood that accumulate in tumours, serves as frontline cells in response to tumour progression owing to the rapid development of micro biotechnology. Hence, targeted therapy with these neutrophils has made targeting treatment a promising field in cancer therapy. MAIN BODY We broadly summarise some studies on the phenotypes and functions of tumour-associated neutrophils as well as the unique web-like products of neutrophils that play a role in cancer progression-neutrophil extracellular traps-and the interactions between neutrophils and the tumour microenvironment. Moreover, several targeted neutrophils therapeutic studies have made some progress and provided potential strategies for the treatment of cancer. CONCLUSION This review aims to offer a holistic perspective on therapeutic interventions targeting neutrophils to further inspire more researches on cancer therapies.
Collapse
Affiliation(s)
- Shuaixi Yang
- Department of Colorectal SurgeryThe First Affiliated Hospital of Zhengzhou UniversityZhengzhou UniversityZhengzhouChina
| | - Jiachi Jia
- Department of Colorectal SurgeryThe First Affiliated Hospital of Zhengzhou UniversityZhengzhou UniversityZhengzhouChina
| | - Fuqi Wang
- Department of Colorectal SurgeryThe First Affiliated Hospital of Zhengzhou UniversityZhengzhou UniversityZhengzhouChina
| | - Yuhang Wang
- Department of Colorectal SurgeryThe First Affiliated Hospital of Zhengzhou UniversityZhengzhou UniversityZhengzhouChina
| | - Yingshuai Fang
- Department of Colorectal SurgeryThe First Affiliated Hospital of Zhengzhou UniversityZhengzhou UniversityZhengzhouChina
| | - Yabing Yang
- Department of Colorectal SurgeryThe First Affiliated Hospital of Zhengzhou UniversityZhengzhou UniversityZhengzhouChina
| | - Quanbo Zhou
- Department of Colorectal SurgeryThe First Affiliated Hospital of Zhengzhou UniversityZhengzhou UniversityZhengzhouChina
| | - Weitang Yuan
- Department of Colorectal SurgeryThe First Affiliated Hospital of Zhengzhou UniversityZhengzhou UniversityZhengzhouChina
| | - Zhilei Bian
- Department of HematologyThe First Affiliated Hospital of Zhengzhou UniversityZhengzhou UniversityZhengzhouChina
| |
Collapse
|
26
|
Dutta S, Sen S. Preparation and Characterization of Collagen-Hyaluronic Acid (Col-HA) Matrices: In Vitro Mimics of the Tumor Microenvironment. Methods Mol Biol 2024; 2747:131-139. [PMID: 38038937 DOI: 10.1007/978-1-0716-3589-6_11] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/02/2023]
Abstract
Collagen-I (Col) and hyaluronic acid (HA) are two of the most important extracellular matrix (ECM) components. While collagen serves as the main structural feature of the ECM and is an important ligand for integrin-based adhesions, HA acts as a ligand for the transmembrane glycoprotein CD44. However, most studies on cancer invasion utilize 3D collagen matrices, thereby ignoring the importance of HA-CD44 interactions. Here, we describe the process of fabricating Col-HA gels with varying Col/HA ratios, which can be used to suitably mimic the tumor microenvironment.
Collapse
Affiliation(s)
- Sarbajeet Dutta
- Department of Biosciences & Bioengineering, IIT Bombay, Mumbai, India
| | - Shamik Sen
- Department of Biosciences & Bioengineering, IIT Bombay, Mumbai, India.
| |
Collapse
|
27
|
Park Y, Chung C. Immune Evasion of G-CSF and GM-CSF in Lung Cancer. Tuberc Respir Dis (Seoul) 2024; 87:22-30. [PMID: 37726942 PMCID: PMC10758314 DOI: 10.4046/trd.2023.0037] [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: 03/23/2023] [Revised: 07/20/2023] [Accepted: 09/12/2023] [Indexed: 09/21/2023] Open
Abstract
Tumor immune evasion is a complex process that involves various mechanisms, such as antigen recognition restriction, immune system suppression, and T cell exhaustion. The tumor microenvironment contains various immune cells involved in immune evasion. Recent studies have demonstrated that granulocyte colony-stimulating factor (G-CSF) and granulocyte-macrophage colony-stimulating factor (GM-CSF) induce immune evasion in lung cancer by modulating neutrophils and myeloid-derived suppressor cells. Here we describe the origin and function of G-CSF and GM-CSF, particularly their role in immune evasion in lung cancer. In addition, their effects on programmed death-ligand 1 expression and clinical implications are discussed.
Collapse
Affiliation(s)
- Yeonhee Park
- Division of Pulmonary and Critical Care Medicine, Department of Internal Medicine, Daejeon St. Mary’s Hospital, College of Medicine, The Catholic University of Korea, Daejeon, Republic of Korea
| | - Chaeuk Chung
- Division of Pulmonology and Critical Care Medicine, Department of Internal Medicine, Chungnam National University College of Medicine, Daejeon, Republic of Korea
- Infection Control Convergence Research Center, Chungnam National University College of Medicine, Daejeon, Republic of Korea
| |
Collapse
|
28
|
Saxena N, Chakraborty S, Dutta S, Bhardwaj G, Karnik N, Shetty O, Jadhav S, Zafar H, Sen S. Stiffness-dependent MSC homing and differentiation into CAFs - implications for breast cancer invasion. J Cell Sci 2024; 137:jcs261145. [PMID: 38108421 DOI: 10.1242/jcs.261145] [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: 03/09/2023] [Accepted: 12/03/2023] [Indexed: 12/19/2023] Open
Abstract
Cellular heterogeneity and extracellular matrix (ECM) stiffening have been shown to be drivers of breast cancer invasiveness. Here, we examine how stiffness-dependent crosstalk between cancer cells and mesenchymal stem cells (MSCs) within an evolving tumor microenvironment regulates cancer invasion. By analyzing previously published single-cell RNA sequencing datasets, we establish the existence of a subpopulation of cells in primary tumors, secondary sites and circulatory tumor cell clusters of highly aggressive triple-negative breast cancer (TNBC) that co-express MSC and cancer-associated fibroblast (CAF) markers. By using hydrogels with stiffnesses of 0.5, 2 and 5 kPa to mimic different stages of ECM stiffening, we show that conditioned medium from MDA-MB-231 TNBC cells cultured on 2 kPa gels, which mimic the pre-metastatic stroma, drives efficient MSC chemotaxis and induces stable differentiation of MSC-derived CAFs in a TGFβ (TGFB1)- and contractility-dependent manner. In addition to enhancing cancer cell proliferation, MSC-derived CAFs on 2 kPa gels maximally boost local invasion and confer resistance to flow-induced shear stresses. Collectively, our results suggest that homing of MSCs at the pre-metastatic stage and their differentiation into CAFs actively drives breast cancer invasion and metastasis in TNBC.
Collapse
Affiliation(s)
- Neha Saxena
- Department of Chemical Engineering, IIT Bombay,Mumbai 400076, India
- Department of Biosciences and Bioengineering, IIT Bombay, Mumbai 400076, India
| | - Soura Chakraborty
- Department of Biological Sciences and Bioengineering, IIT Kanpur, Kanpur 208016, India
| | - Sarbajeet Dutta
- Department of Biosciences and Bioengineering, IIT Bombay, Mumbai 400076, India
| | - Garvit Bhardwaj
- Department of Electrical Engineering, IIT Kanpur, Kanpur 208016, India
| | - Nupur Karnik
- Department of Pathology, Tata Memorial Hospital, Parel, Mumbai 400012, India
| | - Omshree Shetty
- Department of Pathology, Tata Memorial Hospital, Parel, Mumbai 400012, India
| | - Sameer Jadhav
- Department of Chemical Engineering, IIT Bombay,Mumbai 400076, India
| | - Hamim Zafar
- Department of Biological Sciences and Bioengineering, IIT Kanpur, Kanpur 208016, India
- Department of Computer Science and Engineering, IIT Kanpur, Kanpur 208016, India
- Mehta Family Centre for Engineering in Medicine , IIT Kanpur, Kanpur 208016, India
| | - Shamik Sen
- Department of Biosciences and Bioengineering, IIT Bombay, Mumbai 400076, India
| |
Collapse
|
29
|
Lenka S, Bhola RK, Varanasi PR, Bhuyan SK, Bhuyan R. Understanding the functional relevance of oral neutrophils, phenotype and properties in OSCC. Med Oncol 2023; 40:134. [PMID: 37010645 DOI: 10.1007/s12032-023-02010-z] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2023] [Accepted: 03/24/2023] [Indexed: 04/04/2023]
Abstract
Neutrophils are the predominant white blood cells (WBC) that are recruited to the sites of inflammation and infection. They are acknowledged to perform dual roles by promoting (pro-tumor) or by exhibiting anti-cancer properties (anti-tumor). Neutrophils are characterized based on the changes in phenotype and functional properties. To this context, circulating polymorphonuclear neutrophils (cPMN) and tumor-associated neutrophils (TANs) in cancer biology has been well explored but limited to oral polymorphonuclear neutrophils (oPMNs) in oral squamous cell carcinoma (OSCC). However, oPMNs are eminent in maintaining the healthy oral ecosystem by neutralizing microorganisms. Neutralization process enhances the expression of cell surface markers (CD11b, CD63, CD66, CD66b, CD66c, and CD66e) and inflammatory cytokines (TNF-α, IFN-γ, GM-CSF, and IL-8) and increases the recruitment of neutrophils. Along with the inflammation, it has been reported that CEACAM1 and chemerin also favors the infiltration of neutrophils to the cancer site. This indicates that oPMN might contribute to the aetiology of OSCC. The main objective of this review is to explore, the production and migration of oPMNs to the oral cavity, their phenotypes and possible role in OSCC.
Collapse
Affiliation(s)
- Sudhansubala Lenka
- Department of Medical Research, IMS and SUM Hospital, Siksha 'O' Anusandhan Deemed to be University, Bhubaneswar, Odisha, India
| | - Rajesh Kumar Bhola
- Department of Pathology, IMS and SUM Hospital, Siksha 'O' Anusandhan Deemed to be University, Bhubaneswar, Odisha, India
| | - Pavankumar R Varanasi
- Department of Medical Research, IMS and SUM Hospital, Siksha 'O' Anusandhan Deemed to be University, Bhubaneswar, Odisha, India
| | - Sanat Kumar Bhuyan
- Department of Oral Medicine and Radiology, Siksha 'O' Anusandhan Deemed to be University, Bhubaneswar, Odisha, India
| | - Ruchi Bhuyan
- Department of Medical Research, IMS and SUM Hospital, Siksha 'O' Anusandhan Deemed to be University, Bhubaneswar, Odisha, India.
- Department of Oral Pathology and Microbiology, IMS and SUM Hospital, Siksha 'O' Anusandhan (Deemed to be) University, Bhubaneswar, 751003, India.
| |
Collapse
|
30
|
de Visser KE, Joyce JA. The evolving tumor microenvironment: From cancer initiation to metastatic outgrowth. Cancer Cell 2023; 41:374-403. [PMID: 36917948 DOI: 10.1016/j.ccell.2023.02.016] [Citation(s) in RCA: 1360] [Impact Index Per Article: 680.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/20/2022] [Revised: 01/28/2023] [Accepted: 02/14/2023] [Indexed: 03/14/2023]
Abstract
Cancers represent complex ecosystems comprising tumor cells and a multitude of non-cancerous cells, embedded in an altered extracellular matrix. The tumor microenvironment (TME) includes diverse immune cell types, cancer-associated fibroblasts, endothelial cells, pericytes, and various additional tissue-resident cell types. These host cells were once considered bystanders of tumorigenesis but are now known to play critical roles in the pathogenesis of cancer. The cellular composition and functional state of the TME can differ extensively depending on the organ in which the tumor arises, the intrinsic features of cancer cells, the tumor stage, and patient characteristics. Here, we review the importance of the TME in each stage of cancer progression, from tumor initiation, progression, invasion, and intravasation to metastatic dissemination and outgrowth. Understanding the complex interplay between tumor cell-intrinsic, cell-extrinsic, and systemic mediators of disease progression is critical for the rational development of effective anti-cancer treatments.
Collapse
Affiliation(s)
- Karin E de Visser
- Division of Tumor Biology and Immunology, Oncode Institute, The Netherlands Cancer Institute, 1066 CX Amsterdam, the Netherlands; Department of Immunology, Leiden University Medical Center, 2333 ZA Leiden, the Netherlands.
| | - Johanna A Joyce
- Department of Oncology, University of Lausanne, 1011 Lausanne, Switzerland; Ludwig Institute for Cancer Research, 1011 Lausanne, Switzerland; Agora Cancer Center Lausanne, and Swiss Cancer Center Léman, 1011 Lausanne, Switzerland.
| |
Collapse
|
31
|
Nguyen DT, Pedro DI, Pepe A, Rosa JG, Bowman JI, Trachsel L, Golde GR, Suzuki I, Lavrador JM, Nguyen NTY, Kis MA, Smolchek RA, Diodati N, Liu R, Phillpot SR, Webber AR, Castillo P, Sayour EJ, Sumerlin BS, Sawyer WG. Bioconjugation of COL1 protein on liquid-like solid surfaces to study tumor invasion dynamics. Biointerphases 2023; 18:021001. [PMID: 36898958 PMCID: PMC10008099 DOI: 10.1116/6.0002083] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2022] [Revised: 02/02/2023] [Accepted: 02/08/2023] [Indexed: 03/12/2023] Open
Abstract
Tumor invasion is likely driven by the product of intrinsic and extrinsic stresses, reduced intercellular adhesion, and reciprocal interactions between the cancer cells and the extracellular matrix (ECM). The ECM is a dynamic material system that is continuously evolving with the tumor microenvironment. Although it is widely reported that cancer cells degrade the ECM to create paths for migration using membrane-bound and soluble enzymes, other nonenzymatic mechanisms of invasion are less studied and not clearly understood. To explore tumor invasion that is independent of enzymatic degradation, we have created an open three-dimensional (3D) microchannel network using a novel bioconjugated liquid-like solid (LLS) medium to mimic both the tortuosity and the permeability of a loose capillary-like network. The LLS is made from an ensemble of soft granular microgels, which provides an accessible platform to investigate the 3D invasion of glioblastoma (GBM) tumor spheroids using in situ scanning confocal microscopy. The surface conjugation of the LLS microgels with type 1 collagen (COL1-LLS) enables cell adhesion and migration. In this model, invasive fronts of the GBM microtumor protruded into the proximal interstitial space and may have locally reorganized the surrounding COL1-LLS. Characterization of the invasive paths revealed a super-diffusive behavior of these fronts. Numerical simulations suggest that the interstitial space guided tumor invasion by restricting available paths, and this physical restriction is responsible for the super-diffusive behavior. This study also presents evidence that cancer cells utilize anchorage-dependent migration to explore their surroundings, and geometrical cues guide 3D tumor invasion along the accessible paths independent of proteolytic ability.
Collapse
Affiliation(s)
- D. T. Nguyen
- Department of Mechanical and Aerospace Engineering, Herbert Wertheim College of Engineering, College of Medicine University of Florida, Gainesville, Florida 3261
| | - D. I. Pedro
- Department of Mechanical and Aerospace Engineering, Herbert Wertheim College of Engineering, College of Medicine University of Florida, Gainesville, Florida 3261
| | - A. Pepe
- Department of Mechanical and Aerospace Engineering, Herbert Wertheim College of Engineering, College of Medicine University of Florida, Gainesville, Florida 3261
| | - J. G. Rosa
- Department of Mechanical and Aerospace Engineering, Herbert Wertheim College of Engineering, College of Medicine University of Florida, Gainesville, Florida 3261
| | - J. I. Bowman
- Department of Chemistry, College of Liberal Arts and Sciences, College of Medicine University of Florida, Gainesville, Florida 3261
| | - L. Trachsel
- Department of Chemistry, College of Liberal Arts and Sciences, College of Medicine University of Florida, Gainesville, Florida 3261
| | - G. R. Golde
- Department of Mechanical and Aerospace Engineering, Herbert Wertheim College of Engineering, College of Medicine University of Florida, Gainesville, Florida 3261
| | - I. Suzuki
- Department of Mechanical and Aerospace Engineering, Herbert Wertheim College of Engineering, College of Medicine University of Florida, Gainesville, Florida 3261
| | - J. M. Lavrador
- Department of Mechanical and Aerospace Engineering, Herbert Wertheim College of Engineering, College of Medicine University of Florida, Gainesville, Florida 3261
| | - N. T. Y. Nguyen
- Department of Mechanical and Aerospace Engineering, Herbert Wertheim College of Engineering, College of Medicine University of Florida, Gainesville, Florida 3261
| | - M. A. Kis
- Department of Mechanical and Aerospace Engineering, Herbert Wertheim College of Engineering, College of Medicine University of Florida, Gainesville, Florida 3261
| | - R. A. Smolchek
- Department of Mechanical and Aerospace Engineering, Herbert Wertheim College of Engineering, College of Medicine University of Florida, Gainesville, Florida 3261
| | - N. Diodati
- Department of Mechanical and Aerospace Engineering, Herbert Wertheim College of Engineering, College of Medicine University of Florida, Gainesville, Florida 3261
| | - R. Liu
- Department of Surgery, College of Medicine University of Florida, Gainesville, Florida 3261
| | - S. R. Phillpot
- Department of Materials Science and Engineering Herbert Wertheim College of Engineering, College of Medicine University of Florida, Gainesville, Florida 3261
| | - A. R. Webber
- Department of Materials Science and Engineering Herbert Wertheim College of Engineering, College of Medicine University of Florida, Gainesville, Florida 3261
| | - P. Castillo
- Department of Pediatrics, College of Medicine University of Florida, Gainesville, Florida 3261
| | | | - B. S. Sumerlin
- Department of Chemistry, College of Liberal Arts and Sciences, College of Medicine University of Florida, Gainesville, Florida 3261
| | - W. G. Sawyer
- Author to whom correspondence should be addressed:
| |
Collapse
|
32
|
Mierke CT. The versatile roles of ADAM8 in cancer cell migration, mechanics, and extracellular matrix remodeling. Front Cell Dev Biol 2023; 11:1130823. [PMID: 36910158 PMCID: PMC9995898 DOI: 10.3389/fcell.2023.1130823] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2022] [Accepted: 02/15/2023] [Indexed: 02/25/2023] Open
Abstract
The posttranslational proteolytic cleavage is a unique and irreversible process that governs the function and half-life of numerous proteins. Thereby the role of the family of A disintegrin and metalloproteases (ADAMs) plays a leading part. A member of this family, ADAM8, has gained attention in regulating disorders, such as neurogenerative diseases, immune function and cancer, by attenuating the function of proteins nearby the extracellular membrane leaflet. This process of "ectodomain shedding" can alter the turnover rate of a number of transmembrane proteins that function in cell adhesion and receptor signal transduction. In the past, the major focus of research about ADAMs have been on neurogenerative diseases, such as Alzheimer, however, there seems to be evidence for a connection between ADAM8 and cancer. The role of ADAMs in the field of cancer research has gained recent attention, but it has been not yet been extensively addressed. Thus, this review article highlights the various roles of ADAM8 with particular emphasis on pathological conditions, such as cancer and malignant cancer progression. Here, the shedding function, direct and indirect matrix degradation, effects on cancer cell mobility and transmigration, and the interplay of ADAM8 with matrix-embedded neighboring cells are presented and discussed. Moreover, the most probable mechanical impact of ADAM8 on cancer cells and their matrix environment is addressed and debated. In summary, this review presents recent advances in substrates/ligands and functions of ADAM8 in its new role in cancer and its potential link to cell mechanical properties and discusses matrix mechanics modifying properties. A deeper comprehension of the regulatory mechanisms governing the expression, subcellular localization, and activity of ADAM8 is expected to reveal appropriate drug targets that will permit a more tailored and fine-tuned modification of its proteolytic activity in cancer development and metastasis.
Collapse
Affiliation(s)
- Claudia Tanja Mierke
- Faculty of Physics and Earth Science, Biological Physics Division, Peter Debye Institute of Soft Matter Physics, Leipzig University, Leipzig, Germany
| |
Collapse
|
33
|
Zarubova J, Hasani-Sadrabadi MM, Norris SCP, Majedi FS, Xiao C, Kasko AM, Li S. Cell-Taxi: Mesenchymal Cells Carry and Transport Clusters of Cancer Cells. SMALL (WEINHEIM AN DER BERGSTRASSE, GERMANY) 2022; 18:e2203515. [PMID: 36307906 PMCID: PMC9772300 DOI: 10.1002/smll.202203515] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/10/2022] [Revised: 09/09/2022] [Indexed: 06/16/2023]
Abstract
Cell clusters that collectively migrate from primary tumors appear to be far more potent in forming distant metastases than single cancer cells. A better understanding of the collective cell migration phenomenon and the involvement of various cell types during this process is needed. Here, an in vitro platform based on inverted-pyramidal microwells to follow and quantify the collective migration of hundreds of tumor cell clusters at once is developed. These results indicate that mesenchymal stromal cells (MSCs) or cancer-associated fibroblasts (CAFs) in the heterotypic tumor cell clusters may facilitate metastatic dissemination by transporting low-motile cancer cells in a Rac-dependent manner and that extracellular vesicles secreted by mesenchymal cells only play a minor role in this process. Furthermore, in vivo studies show that cancer cell spheroids containing MSCs or CAFs have faster spreading rates. These findings highlight the active role of co-traveling stromal cells in the collective migration of tumor cell clusters and may help in developing better-targeted therapies.
Collapse
Affiliation(s)
- Jana Zarubova
- Department of Bioengineering, University of California, 420 Westwood Plaza, 5121 Engineering V, Los Angeles, CA, 90095-1600, USA
- Department of Biomaterials and Tissue Engineering, Institute of Physiology of the Czech Academy of Sciences, Prague, 14220, Czech Republic
| | - Mohammad Mahdi Hasani-Sadrabadi
- Department of Bioengineering, University of California, 420 Westwood Plaza, 5121 Engineering V, Los Angeles, CA, 90095-1600, USA
| | - Sam C P Norris
- Department of Bioengineering, University of California, 420 Westwood Plaza, 5121 Engineering V, Los Angeles, CA, 90095-1600, USA
| | - Fatemeh Sadat Majedi
- Department of Bioengineering, University of California, 420 Westwood Plaza, 5121 Engineering V, Los Angeles, CA, 90095-1600, USA
| | - Crystal Xiao
- Department of Bioengineering, University of California, 420 Westwood Plaza, 5121 Engineering V, Los Angeles, CA, 90095-1600, USA
| | - Andrea M Kasko
- Department of Bioengineering, University of California, 420 Westwood Plaza, 5121 Engineering V, Los Angeles, CA, 90095-1600, USA
| | - Song Li
- Department of Bioengineering, University of California, 420 Westwood Plaza, 5121 Engineering V, Los Angeles, CA, 90095-1600, USA
| |
Collapse
|
34
|
Dadmehr M, Mortezaei M, Korouzhdehi B. Dual mode fluorometric and colorimetric detection of matrix metalloproteinase MMP-9 as a cancer biomarker based on AuNPs@gelatin/AuNCs nanocomposite. Biosens Bioelectron 2022; 220:114889. [DOI: 10.1016/j.bios.2022.114889] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2022] [Revised: 11/01/2022] [Accepted: 11/04/2022] [Indexed: 11/08/2022]
|
35
|
Kumar L, Bisen M, Khan A, Kumar P, Patel SKS. Role of Matrix Metalloproteinases in Musculoskeletal Diseases. Biomedicines 2022; 10:biomedicines10102477. [PMID: 36289739 PMCID: PMC9598837 DOI: 10.3390/biomedicines10102477] [Citation(s) in RCA: 17] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2022] [Revised: 09/27/2022] [Accepted: 09/30/2022] [Indexed: 11/16/2022] Open
Abstract
Musculoskeletal disorders include rheumatoid arthritis, osteoarthritis, sarcopenia, injury, stiffness, and bone loss. The prevalence of these conditions is frequent among elderly populations with significant mobility and mortality rates. This may lead to extreme discomfort and detrimental effect on the patient’s health and socioeconomic situation. Muscles, ligaments, tendons, and soft tissue are vital for body function and movement. Matrix metalloproteinases (MMPs) are regulatory proteases involved in synthesizing, degrading, and remodeling extracellular matrix (ECM) components. By modulating ECM reconstruction, cellular migration, and differentiation, MMPs preserve myofiber integrity and homeostasis. In this review, the role of MMPs in skeletal muscle function, muscle injury and repair, skeletal muscle inflammation, and muscular dystrophy and future approaches for MMP-based therapies in musculoskeletal disorders are discussed at the cellular and molecule level.
Collapse
Affiliation(s)
- Lokender Kumar
- School of Biotechnology, Faculty of Applied Sciences and Biotechnology, Shoolini University, Solan 173229, India
- Correspondence: (L.K.); (S.K.S.P.); Tel.: +91-017-9235-0000 (L.K.)
| | - Monish Bisen
- School of Biotechnology, Faculty of Applied Sciences and Biotechnology, Shoolini University, Solan 173229, India
| | - Azhar Khan
- School of Biotechnology, Faculty of Applied Sciences and Biotechnology, Shoolini University, Solan 173229, India
| | - Pradeep Kumar
- School of Biotechnology, Faculty of Applied Sciences and Biotechnology, Shoolini University, Solan 173229, India
| | - Sanjay Kumar Singh Patel
- Department of Chemical Engineering, Konkuk University, Seoul 05029, Korea
- Correspondence: (L.K.); (S.K.S.P.); Tel.: +91-017-9235-0000 (L.K.)
| |
Collapse
|
36
|
The Role of Matrix Metalloproteinase in Inflammation with a Focus on Infectious Diseases. Int J Mol Sci 2022; 23:ijms231810546. [PMID: 36142454 PMCID: PMC9500641 DOI: 10.3390/ijms231810546] [Citation(s) in RCA: 52] [Impact Index Per Article: 17.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2022] [Revised: 09/06/2022] [Accepted: 09/08/2022] [Indexed: 11/25/2022] Open
Abstract
Matrix metalloproteinases (MMPs) are involved in extracellular matrix remodeling through the degradation of extracellular matrix components and are also involved in the inflammatory response by regulating the pro-inflammatory cytokines TNF-α and IL-1β. Dysregulation in the inflammatory response and changes in the extracellular matrix by MMPs are related to the development of various diseases including lung and cardiovascular diseases. Therefore, numerous studies have been conducted to understand the role of MMPs in disease pathogenesis. MMPs are involved in the pathogenesis of infectious diseases through a dysregulation of the activity and expression of MMPs. In this review, we discuss the role of MMPs in infectious diseases and inflammatory responses. Furthermore, we present the potential of MMPs as therapeutic targets in infectious diseases.
Collapse
|
37
|
Martinez-Garcia FD, van Dongen JA, Burgess JK, Harmsen MC. Matrix Metalloproteases from Adipose Tissue-Derived Stromal Cells Are Spatiotemporally Regulated by Hydrogel Mechanics in a 3D Microenvironment. BIOENGINEERING (BASEL, SWITZERLAND) 2022; 9:bioengineering9080340. [PMID: 35892753 PMCID: PMC9332414 DOI: 10.3390/bioengineering9080340] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/10/2022] [Revised: 07/11/2022] [Accepted: 07/16/2022] [Indexed: 01/16/2023]
Abstract
Adipose tissue-derived stromal cells (ASCs) are of interest in tissue engineering and regenerative medicine (TERM) due to their easy acquisition, multipotency, and secretion of a host of factors that promote regeneration. Retention of ASCs in or around lesions is poor following direct administration. Therefore, for TERM applications, ASCs can be ‘immobilized’ via their incorporation into hydrogels such as gelatine methacryloyl (GelMA). Tweaking GelMA concentration is a common approach to approximate the mechanical properties found in organs or tissues that need repair. Distinct hydrogel mechanics influence the ability of a cell to spread, migrate, proliferate, and secrete trophic factors. Mesenchymal cells such as ASCs are potent remodellers of the extracellular matrix (ECM). Not only do ASCs deposit components, they also secrete matrix metalloproteases (MMPs) which degrade ECM. In this work, we investigated if GelMA polymer concentration influenced the expression of active MMPs by ASCs. In addition, MMPs’ presence was interrogated with regard to ASCs morphology and changes in hydrogel ultrastructure. For this, immortalised ASCs were embedded in 5%, 10%, and 15% (w/v) GelMA hydrogels, photopolymerised and cultured for 14 d. Zymography in situ indicated that MMPs had a variable, hydrogel concentration-dependent influence on ASCs-secreted MMPs. In 5% GelMA, ASCs showed a high and sustained expression of MMPs, while, in 10% and 15% GelMA, such expression was almost null. ASCs morphology based on F-actin staining showed that increasing GelMA concentrations inhibit their spreading. Scanning electron microscopy (SEM) showed that hydrogel ultrastructure in terms of pore density, pore size, and percentage porosity were not consistently influenced by cells. Interestingly, changes in ultrastructural parameters were detected also in cell-free materials, albeit without a clear trend. We conclude that hydrogel concentration and its underlying mechanics influenced MMP expression by ASCs. The exact MMPs that respond to these mechanical cues should be defined in follow-up experiments.
Collapse
Affiliation(s)
- Francisco Drusso Martinez-Garcia
- Department of Pathology and Medical Biology, University Medical Center Groningen, University of Groningen, 9713 GZ Groningen, The Netherlands; (F.D.M.-G.); (J.K.B.)
- W.J. Kolff Research Institute, University Medical Center Groningen, University of Groningen, 9713 GZ Groningen, The Netherlands
| | - Joris Anton van Dongen
- Department of Plastic Surgery, University Medical Center Utrecht, 3584 CX Utrecht, The Netherlands;
| | - Janette Kay Burgess
- Department of Pathology and Medical Biology, University Medical Center Groningen, University of Groningen, 9713 GZ Groningen, The Netherlands; (F.D.M.-G.); (J.K.B.)
- W.J. Kolff Research Institute, University Medical Center Groningen, University of Groningen, 9713 GZ Groningen, The Netherlands
- Groningen Research Institute for Asthma and COPD (GRIAC), University Medical Center Groningen, University of Groningen, 9713 GZ Groningen, The Netherlands
| | - Martin Conrad Harmsen
- Department of Pathology and Medical Biology, University Medical Center Groningen, University of Groningen, 9713 GZ Groningen, The Netherlands; (F.D.M.-G.); (J.K.B.)
- W.J. Kolff Research Institute, University Medical Center Groningen, University of Groningen, 9713 GZ Groningen, The Netherlands
- Groningen Research Institute for Asthma and COPD (GRIAC), University Medical Center Groningen, University of Groningen, 9713 GZ Groningen, The Netherlands
- Correspondence:
| |
Collapse
|
38
|
Yun EJ, Kim D, Hsieh JT, Baek ST. Stanniocalcin 2 drives malignant transformation of human glioblastoma cells by targeting SNAI2 and Matrix Metalloproteinases. Cell Death Dis 2022; 8:308. [PMID: 35790735 PMCID: PMC9256701 DOI: 10.1038/s41420-022-01090-6] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2022] [Revised: 06/07/2022] [Accepted: 06/13/2022] [Indexed: 11/26/2022]
Abstract
Glioblastoma multiforme (GBM) is the most malignant brain tumor and is refractory to conventional therapies. Although previous studies have proposed that the interaction between gene mutations and the external environment leads to the occurrence of GBM, the pathogenesis of GBM is still unclear and much remains to be studied. Herein, we show an association between human glycoprotein stanniocalcin-2 (STC2) and aggressive GBM progression, and demonstrate the underlying mechanism. Elevated STC2 expression and secretion greatly increase GBM cell growth and invasive phenotypes. Mechanistically, both, conditioned media (CM) containing STC2 and recombinant STC2, can induce the transformation of GBM cells into more malignant phenotypes by upregulating the expression of the epithelial-mesenchymal transition transcription factor, snail family transcription repressor 2 (SNAI2) as well as matrix metalloproteinases (MMPs). Moreover, we further demonstrate that the oncogenic function of STC2 in GBM is mediated through the MAPK signaling pathway. Collectively, these results identify the mechanism of STC2 targeting SNAI2 and MMPs through the MAPK pathway in GBM, and provide insights into a potential therapeutic strategy for GBM.
Collapse
|
39
|
Kwak SB, Kim SJ, Kim J, Kang YL, Ko CW, Kim I, Park JW. Tumor regionalization after surgery: Roles of the tumor microenvironment and neutrophil extracellular traps. EXPERIMENTAL & MOLECULAR MEDICINE 2022; 54:720-729. [PMID: 35764882 PMCID: PMC9256747 DOI: 10.1038/s12276-022-00784-2] [Citation(s) in RCA: 39] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 01/16/2022] [Revised: 03/20/2022] [Accepted: 03/30/2022] [Indexed: 11/09/2022]
Abstract
Surgery is unanimously regarded as the primary strategy to cure solid tumors in the early stages but is not always used in advanced cases. However, tumor surgery must be carefully considered because the risk of metastasis could be increased by the surgical procedure. Tumor surgery may result in a deep wound, which induces many biological responses favoring tumor metastasis. In particular, NETosis, which is the process of forming neutrophil extracellular traps (NETs), has received attention as a risk factor for surgery-induced metastasis. To reduce cancer mortality, researchers have made efforts to prevent secondary metastasis after resection of the primary tumor. From this point of view, a better understanding of surgery-induced metastasis might provide new strategies for more effective and safer surgical approaches. In this paper, recent insights into the surgical effects on metastasis will be reviewed. Moreover, in-depth opinions about the effects of NETs on metastasis will be discussed. Therapies that limit the formation of web-like structures formed by white cells known as neutrophils may lower the risk of cancer spread (metastasis) following surgical tumor removal. Removing solid tumors remains a key cancer treatment, but in some cases surgery itself increases the risk of metastasis. Jong-Wan Park at Seoul National University, South Korea, and co-workers reviewed current understanding of metastasis following surgery. Surgical removal destroys the architecture supporting cancer cells but this can release tumor cells into blood vessels. The stress of deep wounds also affects immune responses, most notably neutrophil extracellular traps (NETs), web-like structures formed by neutrophils to trap and kill pathogens. NETs have previously been implicated in metastasis. In a post-surgical environment enriched in neutrophils and pro-inflammatory cytokines, NET formation may help cancer cells thrive, promoting metastasis.
Collapse
Affiliation(s)
- Su-Bin Kwak
- Department of Pharmacology, Seoul National University College of Medicine, Daehak-ro, Jongno-gu, Seoul, 03080, Korea.,Department of Biomedical Science, BK21-plus Education Program, Seoul National University College of Medicine, Daehak-ro, Jongno-gu, Seoul, 03080, Korea
| | - Sang Jin Kim
- Department of Pharmacology, Seoul National University College of Medicine, Daehak-ro, Jongno-gu, Seoul, 03080, Korea.,Cancer Research Institute and Ischemic/Hypoxic Disease Institute, Seoul National University College of Medicine, Daehak-ro, Jongno-gu, Seoul, 03080, Korea
| | - Jiyoung Kim
- Department of Pharmacology, Seoul National University College of Medicine, Daehak-ro, Jongno-gu, Seoul, 03080, Korea
| | - Ye-Lim Kang
- Department of Pharmacology, Seoul National University College of Medicine, Daehak-ro, Jongno-gu, Seoul, 03080, Korea.,Department of Biomedical Science, BK21-plus Education Program, Seoul National University College of Medicine, Daehak-ro, Jongno-gu, Seoul, 03080, Korea
| | - Chang Woo Ko
- Department of Pharmacology, Seoul National University College of Medicine, Daehak-ro, Jongno-gu, Seoul, 03080, Korea.,Department of Biomedical Science, BK21-plus Education Program, Seoul National University College of Medicine, Daehak-ro, Jongno-gu, Seoul, 03080, Korea
| | - Iljin Kim
- Department of Pharmacology, Inha University College of Medicine, Inha-ro, Michuhol-gu, Incheon, 22212, Korea
| | - Jong-Wan Park
- Department of Pharmacology, Seoul National University College of Medicine, Daehak-ro, Jongno-gu, Seoul, 03080, Korea. .,Department of Biomedical Science, BK21-plus Education Program, Seoul National University College of Medicine, Daehak-ro, Jongno-gu, Seoul, 03080, Korea. .,Cancer Research Institute and Ischemic/Hypoxic Disease Institute, Seoul National University College of Medicine, Daehak-ro, Jongno-gu, Seoul, 03080, Korea.
| |
Collapse
|
40
|
He K, Liu X, Hoffman RD, Shi RZ, Lv GY, Gao JL. G-CSF/GM-CSF-induced hematopoietic dysregulation in the progression of solid tumors. FEBS Open Bio 2022; 12:1268-1285. [PMID: 35612789 PMCID: PMC9249339 DOI: 10.1002/2211-5463.13445] [Citation(s) in RCA: 27] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2022] [Revised: 05/17/2022] [Accepted: 05/24/2022] [Indexed: 11/06/2022] Open
Abstract
There are two types of abnormal hematopoiesis in solid tumor occurrence and treatment: pathological hematopoiesis, and myelosuppression induced by radiotherapy and chemotherapy. In this review, we primarily focus on the abnormal pathological hematopoietic differentiation in cancer induced by tumor-released granulocyte colony stimulating factor (G-CSF) and granulocyte-macrophage colony stimulating factor (GM-CSF). As key factors in hematopoietic development, G-CSF/GM-CSF are well-known facilitators of myelopoiesis and mobilization of hematopoietic stem cells (HSCs). In addition, these two cytokines can also promote or inhibit tumors, dependent on tumor type. In multiple cancer types, hematopoiesis is greatly enhanced and abnormal lineage differentiation is induced by these two cytokines. Here, dysregulated hematopoiesis induced by G-CSF/GM-CSF in solid tumors and its mechanism are summarized, and the prognostic value of G-CSF/GM-CSF-associated dysregulated hematopoiesis for tumor metastasis is also briefly highlighted.
Collapse
Affiliation(s)
- Kai He
- The First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, Zhejiang, 310009, China
| | - Xi Liu
- School of Basic Medical Sciences, Zhejiang Chinese Medical University, Hangzhou, Zhejiang, 310053, China
| | - Robert D Hoffman
- Yo San University of Traditional Chinese Medicine, Los Angeles, CA, 90066, USA
| | - Rong-Zhen Shi
- Tangqi Branch of Traditional Chinese Medicine Hospital of Yuhang District, Hangzhou, Zhejiang, 311106, China
| | - Gui-Yuan Lv
- School of Pharmaceutical Sciences, Zhejiang Chinese Medical University Hangzhou, Zhejiang, 310053, China
| | - Jian-Li Gao
- School of Pharmaceutical Sciences, Zhejiang Chinese Medical University Hangzhou, Zhejiang, 310053, China
| |
Collapse
|
41
|
Silibinin exerts anti-cancer activity on human ovarian cancer cells by increasing apoptosis and inhibiting epithelial-mesenchymal transition (EMT). Gene 2022; 823:146275. [PMID: 35189245 DOI: 10.1016/j.gene.2022.146275] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2021] [Revised: 01/13/2022] [Accepted: 02/03/2022] [Indexed: 01/14/2023]
Abstract
BACKGROUND Silibinin, the principal flavonoid derived from milk thistle seeds, has been demonstrated to have strong inhibitory effects against human malignancies. The inhibitory function of silibinin on ovarian cancer, however, is not fully identified. In this essay, both in vivo and in vitro investigations were conducted to survey the silibinin's blocking effects on ovarian cancer. METHODS The impacts of silibinin on two ovarian cancer cell lines, SKOV-3 and A2870, were determined by evaluating cell viability, migration, invasion, and apoptosis. Q-RT-PCR and western blotting techniques were carried out to explore the protein levels of signaling pathway markers. A mouse xenograft model was utilized to determine the silibinin efficacy in inhibiting tumor growth. RESULTS After cell treatment with silibinin, cell viability, migration, and invasion were appreciably inhibited in cancer cell lines, but cell apoptosis was promoted. Also, silibinin reversed the epithelial-mesenchymal transition (EMT) mechanism by inducing E-cadherin expression and reducing N-cadherin and vimentin expression, suppressing the levels of regulators related to EMT such as Snail, Slug, and ZEB1 transcription factors, and also decreasing PI3K/AKT, Smad2/3, and β-catenin intermediate molecules in vitro. Silibinin effectively ameliorated tumor growth in vivo. CONCLUSION silibinin could be considered a potent agent against ovarian cancer based on the results.
Collapse
|
42
|
Pally D, Goutham S, Bhat R. Extracellular matrix as a driver for intratumoral heterogeneity. Phys Biol 2022; 19. [PMID: 35545075 DOI: 10.1088/1478-3975/ac6eb0] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2021] [Accepted: 05/11/2022] [Indexed: 11/12/2022]
Abstract
The architecture of an organ is built through interactions between its native cells and its connective tissue consisting of stromal cells and the extracellular matrix (ECM). Upon transformation through tumorigenesis, such interactions are disrupted and replaced by a new set of intercommunications between malignantly transformed parenchyma, an altered stromal cell population, and a remodeled ECM. In this perspective, we propose that the intratumoral heterogeneity of cancer cell phenotypes is an emergent property of such reciprocal intercommunications, both biochemical and mechanical-physical, which engender and amplify the diversity of cell behavioral traits. An attempt to assimilate such findings within a framework of phenotypic plasticity furthers our understanding of cancer progression.
Collapse
Affiliation(s)
- Dharma Pally
- Molecular Reproduction Development and Genetics, Indian Institute of Science, GA 07, Bangalore, Karnataka, 560012, INDIA
| | - Shyamili Goutham
- Molecular Reproduction Development and Genetics, Indian Institute of Science, GA 07, Bangalore, Karnataka, 560012, INDIA
| | - Ramray Bhat
- Molecular Reproduction Development and Genetics, Indian Institute of Science, GA 07, Bangalore, Karnataka, 560012, INDIA
| |
Collapse
|
43
|
Mahmud Z, Rahman A, Mishu ID, Kabir Y. Mechanistic insights into the interplays between neutrophils and other immune cells in cancer development and progression. Cancer Metastasis Rev 2022; 41:405-432. [PMID: 35314951 DOI: 10.1007/s10555-022-10024-8] [Citation(s) in RCA: 23] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/19/2021] [Accepted: 03/09/2022] [Indexed: 12/12/2022]
Abstract
Cancer is considered a major public health concern worldwide and is characterized by an uncontrolled division of abnormal cells. The human immune system recognizes cancerous cells and induces innate immunity to destroy those cells. However, sustained tumors may protect themselves by developing immune escape mechanisms through multiple soluble and cellular mediators. Neutrophils are the most plenteous leukocytes in the human blood and are crucial for immune defense in infection and inflammation. Besides, neutrophils emancipate the antimicrobial contents, secrete different cytokines or chemokines, and interact with other immune cells to combat and successfully kill cancerous cells. Conversely, many clinical and experimental studies signpost that being a polarized and heterogeneous population with plasticity, neutrophils, particularly their subpopulations, act as a modulator of cancer development by promoting tumor metastasis, angiogenesis, and immunosuppression. Studies also suggest that tumor infiltrating macrophages, neutrophils, and other innate immune cells support tumor growth and survival. Additionally, neutrophils promote tumor cell invasion, migration and intravasation, epithelial to mesenchymal transition, survival of cancer cells in the circulation, seeding, and extravasation of tumor cells, and advanced growth and development of cancer cells to form metastases. In this manuscript, we describe and review recent studies on the mechanisms for neutrophil recruitment, activation, and their interplay with different immune cells to promote their pro-tumorigenic functions. Understanding the detailed mechanisms of neutrophil-tumor cell interactions and the concomitant roles of other immune cells will substantially improve the clinical utility of neutrophils in cancer and eventually may aid in the identification of biomarkers for cancer prognosis and the development of novel therapeutic approaches for cancer treatment.
Collapse
Affiliation(s)
- Zimam Mahmud
- Department of Biochemistry and Molecular Biology, University of Dhaka, Dhaka, 1000, Bangladesh
| | - Atiqur Rahman
- Department of Biochemistry and Molecular Biology, University of Dhaka, Dhaka, 1000, Bangladesh
| | | | - Yearul Kabir
- Department of Biochemistry and Molecular Biology, University of Dhaka, Dhaka, 1000, Bangladesh.
| |
Collapse
|
44
|
de los Reyes AA, Kim Y. Optimal regulation of tumour-associated neutrophils in cancer progression. ROYAL SOCIETY OPEN SCIENCE 2022; 9:210705. [PMID: 35127110 PMCID: PMC8808100 DOI: 10.1098/rsos.210705] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 06/10/2021] [Accepted: 11/19/2021] [Indexed: 06/14/2023]
Abstract
In a tumour microenvironment, tumour-associated neutrophils could display two opposing differential phenotypes: anti-tumour (N1) and pro-tumour (N2) effector cells. Converting N2 to N1 neutrophils provides innovative therapies for cancer treatment. In this study, a mathematical model for N1-N2 dynamics describing the cancer survival and immune inhibition in response to TGF-β and IFN-β is considered. The effects of exogenous intervention of TGF-β inhibitor and IFN-β are examined in order to enhance N1 recruitment to combat tumour progression. Our approach employs optimal control theory to determine drug infusion protocols that could minimize tumour volume with least administration cost possible. Four optimal control scenarios corresponding to different therapeutic strategies are explored, namely, TGF-β inhibitor control only, IFN-β control only, concomitant TGF-β inhibitor and IFN-β controls, and alternating TGF-β inhibitor and IFN-β controls. For each scheme, different initial conditions are varied to depict different pathophysiological condition of a cancer patient, leading to adaptive treatment schedule. TGF-β inhibitor and IFN-β drug dosages, total drug amount, infusion times and relative cost of drug administrations are obtained under various circumstances. The control strategies achieved could guide in designing individualized therapeutic protocols.
Collapse
Affiliation(s)
- Aurelio A. de los Reyes
- Biomedical Mathematics Group, Pioneer Research Center for Mathematical and Computational Sciences, Institute for Basic Science, Daejeon 34126, Republic of Korea
- Institute of Mathematics, University of the Philippines Diliman, Quezon City 1101, Philippines
| | - Yangjin Kim
- Department of Mathematics, Konkuk University, Seoul 05029, Republic of Korea
- Mathematical Biosciences Institute, Columbus, OH 43210, USA
| |
Collapse
|
45
|
Nisiewicz MK, Gajda A, Kowalczyk A, Cupriak A, Kasprzak A, Bamburowicz-Klimkowska M, Grudzinski IP, Nowicka AM. Novel electrogravimetric biosensors for the ultrasensitive detection of plasma matrix metalloproteinase-2 considered a potential tumor biomarker. Anal Chim Acta 2022; 1191:339290. [PMID: 35033237 DOI: 10.1016/j.aca.2021.339290] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2021] [Revised: 10/29/2021] [Accepted: 11/15/2021] [Indexed: 11/17/2022]
Abstract
In this study, we developed novel, simple gravimetric and voltammetric sensors for the ultrasensitive detection of active matrix metalloproteinase (MMP)-2 in plasma. The developed sensors are cost-effective, require a very less amount of reagents, and are time-saving. They detect MMP-2 based on antigen-antibody recognition and its ability to cleave glycine-leucine peptide bond. The three-dimensional bioplatform of the sensors consisted of a cationic polyethyleneimine (PEI) polymer that facilitated robust immobilization of the dipeptide labeled with anthraquinone (AQ), or antibody molecules in appropriate density, which was crucial for biosensing. Detection was performed using quartz crystal microbalance with dissipation and voltammetry. The results showed that the developed sensors were characterized by high stability, wide analytical range (2.0 pg mL-1 to 5.0 μg mL-1), and low detection limit (ca. 10 fg mL-1). They also exhibited excellent efficiency in the determination of active MMP-2 in real samples, such as blood plasma. The developed sensors may hold great promise for the early diagnosis of cancers.
Collapse
Affiliation(s)
- Monika K Nisiewicz
- Faculty of Chemistry, University of Warsaw, Pasteura Str. 1, PL 02-093, Warsaw, Poland; Faculty of Chemistry, Warsaw University of Technology, Noakowskiego Str. 3, PL 00-664, Warsaw, Poland
| | - Aleksandra Gajda
- Faculty of Chemistry, University of Warsaw, Pasteura Str. 1, PL 02-093, Warsaw, Poland
| | - Agata Kowalczyk
- Faculty of Chemistry, University of Warsaw, Pasteura Str. 1, PL 02-093, Warsaw, Poland
| | - Aleksandra Cupriak
- Faculty of Chemistry, University of Warsaw, Pasteura Str. 1, PL 02-093, Warsaw, Poland
| | - Artur Kasprzak
- Faculty of Chemistry, Warsaw University of Technology, Noakowskiego Str. 3, PL 00-664, Warsaw, Poland
| | | | - Ireneusz P Grudzinski
- Faculty of Pharmacy, Medical University of Warsaw, Banacha Str. 1, PL-02-097, Warsaw, Poland
| | - Anna M Nowicka
- Faculty of Chemistry, University of Warsaw, Pasteura Str. 1, PL 02-093, Warsaw, Poland.
| |
Collapse
|
46
|
Cao H, Duan L, Zhang Y, Cao J, Zhang K. Current hydrogel advances in physicochemical and biological response-driven biomedical application diversity. Signal Transduct Target Ther 2021; 6:426. [PMID: 34916490 PMCID: PMC8674418 DOI: 10.1038/s41392-021-00830-x] [Citation(s) in RCA: 431] [Impact Index Per Article: 107.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2021] [Revised: 11/10/2021] [Accepted: 11/11/2021] [Indexed: 02/05/2023] Open
Abstract
Hydrogel is a type of versatile platform with various biomedical applications after rational structure and functional design that leverages on material engineering to modulate its physicochemical properties (e.g., stiffness, pore size, viscoelasticity, microarchitecture, degradability, ligand presentation, stimulus-responsive properties, etc.) and influence cell signaling cascades and fate. In the past few decades, a plethora of pioneering studies have been implemented to explore the cell-hydrogel matrix interactions and figure out the underlying mechanisms, paving the way to the lab-to-clinic translation of hydrogel-based therapies. In this review, we first introduced the physicochemical properties of hydrogels and their fabrication approaches concisely. Subsequently, the comprehensive description and deep discussion were elucidated, wherein the influences of different hydrogels properties on cell behaviors and cellular signaling events were highlighted. These behaviors or events included integrin clustering, focal adhesion (FA) complex accumulation and activation, cytoskeleton rearrangement, protein cyto-nuclei shuttling and activation (e.g., Yes-associated protein (YAP), catenin, etc.), cellular compartment reorganization, gene expression, and further cell biology modulation (e.g., spreading, migration, proliferation, lineage commitment, etc.). Based on them, current in vitro and in vivo hydrogel applications that mainly covered diseases models, various cell delivery protocols for tissue regeneration and disease therapy, smart drug carrier, bioimaging, biosensor, and conductive wearable/implantable biodevices, etc. were further summarized and discussed. More significantly, the clinical translation potential and trials of hydrogels were presented, accompanied with which the remaining challenges and future perspectives in this field were emphasized. Collectively, the comprehensive and deep insights in this review will shed light on the design principles of new biomedical hydrogels to understand and modulate cellular processes, which are available for providing significant indications for future hydrogel design and serving for a broad range of biomedical applications.
Collapse
Affiliation(s)
- Huan Cao
- Department of Nuclear Medicine, West China Hospital, and National Engineering Research Center for Biomaterials, Sichuan University, 610064, Chengdu, P. R. China
- Department of Medical Ultrasound and Central Laboratory, Shanghai Tenth People's Hospital, Tongji University School of Medicine, No. 301 Yan-chang-zhong Road, 200072, Shanghai, People's Republic of China
- School of Materials Science and Engineering, Nanyang Technological University, 50 Nanyang Avenue, Singapore, 639798, Singapore
| | - Lixia Duan
- Department of Medical Ultrasound and Central Laboratory, Shanghai Tenth People's Hospital, Tongji University School of Medicine, No. 301 Yan-chang-zhong Road, 200072, Shanghai, People's Republic of China
| | - Yan Zhang
- Department of Medical Ultrasound and Central Laboratory, Shanghai Tenth People's Hospital, Tongji University School of Medicine, No. 301 Yan-chang-zhong Road, 200072, Shanghai, People's Republic of China
| | - Jun Cao
- Department of Nuclear Medicine, West China Hospital, and National Engineering Research Center for Biomaterials, Sichuan University, 610064, Chengdu, P. R. China.
| | - Kun Zhang
- Department of Medical Ultrasound and Central Laboratory, Shanghai Tenth People's Hospital, Tongji University School of Medicine, No. 301 Yan-chang-zhong Road, 200072, Shanghai, People's Republic of China.
| |
Collapse
|
47
|
Dieffenbach PB, Mallarino Haeger C, Rehman R, Corcoran AM, Coronata AMF, Vellarikkal SK, Chrobak I, Waxman AB, Vitali SH, Sholl LM, Padera RF, Lagares D, Polverino F, Owen CA, Fredenburgh LE. A Novel Protective Role for Matrix Metalloproteinase-8 in the Pulmonary Vasculature. Am J Respir Crit Care Med 2021; 204:1433-1451. [PMID: 34550870 PMCID: PMC8865706 DOI: 10.1164/rccm.202108-1863oc] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2021] [Accepted: 09/22/2021] [Indexed: 11/16/2022] Open
Abstract
Rationale: Mechanical signaling through cell-matrix interactions plays a major role in progressive vascular remodeling in pulmonary arterial hypertension (PAH). MMP-8 (matrix metalloproteinase-8) is an interstitial collagenase involved in regulating inflammation and fibrosis of the lung and systemic vasculature, but its role in PAH pathogenesis remains unexplored. Objectives: To evaluate MMP-8 as a modulator of pathogenic mechanical signaling in PAH. Methods: MMP-8 levels were measured in plasma from patients with pulmonary hypertension (PH) and controls by ELISA. MMP-8 vascular expression was examined in lung tissue from patients with PAH and rodent models of PH. MMP-8-/- and MMP-8+/+ mice were exposed to normobaric hypoxia or normoxia for 4-8 weeks. PH severity was evaluated by right ventricular systolic pressure, echocardiography, pulmonary artery morphometry, and immunostaining. Proliferation, migration, matrix component expression, and mechanical signaling were assessed in MMP-8-/- and MMP-8+/+ pulmonary artery smooth muscle cells (PASMCs). Measurements and Main Results: MMP-8 expression was significantly increased in plasma and pulmonary arteries of patients with PH compared with controls and induced in the pulmonary vasculature in rodent PH models. Hypoxia-exposed MMP-8-/- mice had significant mortality, increased right ventricular systolic pressure, severe right ventricular dysfunction, and exaggerated vascular remodeling compared with MMP-8+/+ mice. MMP-8-/- PASMCs demonstrated exaggerated proliferation and migration mediated by altered matrix protein expression, elevated integrin-β3 levels, and induction of FAK (focal adhesion kinase) and downstream YAP (Yes-associated protein)/TAZ (transcriptional coactivator with PDZ-binding motif) activity. Conclusions: MMP-8 is a novel protective factor upregulated in the pulmonary vasculature during PAH pathogenesis. MMP-8 opposes pathologic mechanobiological feedback by altering matrix composition and disrupting integrin-β3/FAK and YAP/TAZ-dependent mechanical signaling in PASMCs.
Collapse
Affiliation(s)
| | | | | | | | | | | | - Izabela Chrobak
- Lovelace Respiratory Research Institute, Albuquerque, New Mexico
| | | | - Sally H. Vitali
- Department of Anesthesiology, Critical Care, and Pain Medicine, Boston Children’s Hospital, Boston, Massachusetts; and
| | - Lynette M. Sholl
- Department of Pathology, Brigham and Women’s Hospital, Boston, Massachusetts
| | - Robert F. Padera
- Department of Pathology, Brigham and Women’s Hospital, Boston, Massachusetts
| | - David Lagares
- Division of Pulmonary and Critical Care Medicine, Massachusetts General Hospital, Boston, Massachusetts
| | | | | | | |
Collapse
|
48
|
Paruchuri BC, Gopal V, Sarupria S, Larsen J. Toward enzyme-responsive polymersome drug delivery. Nanomedicine (Lond) 2021; 16:2679-2693. [PMID: 34870451 DOI: 10.2217/nnm-2021-0194] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023] Open
Abstract
In drug delivery, enzyme-responsive drug carriers are becoming increasingly relevant because of the growing association of disease pathology with enzyme overexpression. Polymersomes are of interest to such applications because of their tunable properties. While polymersomes open up a wide range of chemical and physical properties to explore, they also present a challenge in developing generalized rules for the synthesis of novel systems. Motivated by this issue, in this perspective, we summarize the existing knowledge on enzyme-responsive polymersomes and outline the main design choices. Then, we propose heuristics to guide the design of novel systems. Finally, we discuss the potential of an integrated approach using computer simulations and experimental studies to streamline this design process and close the existing knowledge gaps.
Collapse
Affiliation(s)
| | - Varun Gopal
- Department of Chemical & Biomolecular Engineering, Clemson University, Clemson, SC 29631, USA.,Department of Chemical Engineering & Material Science, University of Minnesota, Minneapolis, MN 55455, USA
| | - Sapna Sarupria
- Department of Chemical & Biomolecular Engineering, Clemson University, Clemson, SC 29631, USA.,Center for Optical Materials Science & Engineering Technologies (COMSET), Clemson University, Clemson, SC 29670, USA.,Department of Chemistry, University of Minnesota, Minneapolis, MN 55455, USA
| | - Jessica Larsen
- Department of Chemical & Biomolecular Engineering, Clemson University, Clemson, SC 29631, USA.,Department of Bioengineering, Clemson University, Clemson, SC 29631, USA
| |
Collapse
|
49
|
Sthanam LK, Roy T, Patwardhan S, Shukla A, Sharma S, Shinde PV, Kale HT, Chandra Shekar P, Kondabagil K, Sen S. MMP modulated differentiation of mouse embryonic stem cells on engineered cell derived matrices. Biomaterials 2021; 280:121268. [PMID: 34871878 DOI: 10.1016/j.biomaterials.2021.121268] [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: 11/15/2019] [Revised: 10/27/2021] [Accepted: 11/17/2021] [Indexed: 11/16/2022]
Abstract
Stem cell differentiation is dictated by the dynamic crosstalk between cells and their underlying extracellular matrix. While the importance of matrix degradation mediated by enzymes such as matrix metalloproteinases (MMPs) in the context of cancer invasion is well established, the role of MMPs in stem cell differentiation remains relatively unexplored. Here we address this question by assaying MMP expression and activity during differentiation of mouse embryonic stem cells (mESCs) on mouse embryonic fibroblast (MEF) derived matrices (MEFDMs) of varying stiffness and composition. We show that mESC differentiation into different germ layers is associated with expression of several MMPs including MMP-11, 2, 17, 25 and 9, with MMP-9 detected in cell secreted media. Different extents of softening of the different MEFDMs led to altered integrin expression, activated distinct mechanotransduction and metabolic pathways, and induced expression of germ layer-specific markers. Inhibition of MMP proteolytic activity by the broad spectrum MMP inhibitor GM6001 led to alterations in germ layer commitment of the differentiating mESCs. Together, our results illustrate the effect of MMPs in regulating mESC differentiation on engineered cell derived matrices and establish MEFDMs as suitable substrates for understanding molecular mechanisms regulating stem cell development and for regenerative medicine applications.
Collapse
Affiliation(s)
| | - Tanusri Roy
- Department. of Biosciences & Bioengineering, IIT Bombay, Mumbai, India
| | - Sejal Patwardhan
- Department. of Biosciences & Bioengineering, IIT Bombay, Mumbai, India; Advanced Centre for Treatment, Research and Education in Cancer - Tata Memorial Centre (ACTREC-TMC), Kharghar, Navi Mumbai, India; Homi Bhabha National Institute, Anushakti Nagar, Mumbai, India
| | - Avi Shukla
- Department. of Biosciences & Bioengineering, IIT Bombay, Mumbai, India
| | - Shipra Sharma
- Department. of Biosciences & Bioengineering, IIT Bombay, Mumbai, India
| | - Pradip V Shinde
- Department. of Biosciences & Bioengineering, IIT Bombay, Mumbai, India
| | | | | | - Kiran Kondabagil
- Department. of Biosciences & Bioengineering, IIT Bombay, Mumbai, India
| | - Shamik Sen
- Department. of Biosciences & Bioengineering, IIT Bombay, Mumbai, India.
| |
Collapse
|
50
|
ECM stiffness-tuned exosomes drive breast cancer motility through thrombospondin-1. Biomaterials 2021; 279:121185. [PMID: 34808560 DOI: 10.1016/j.biomaterials.2021.121185] [Citation(s) in RCA: 90] [Impact Index Per Article: 22.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2021] [Revised: 09/08/2021] [Accepted: 10/12/2021] [Indexed: 12/15/2022]
Abstract
Breast cancer progression features ECM stiffening due to excess deposition and crosslinking of collagen, which dramatically influence tumor behaviour and fate. The mechanisms by which extracellular matrix (ECM) stiffening drives breast cancer invasion is an area of active research. Here we demonstrate the role of exosomes in ECM stiffness triggered breast cancer invasiveness. Using stiffness tuneable hydrogel ECM scaffolds, we show that stiff ECMs promote exosome secretion in a YAP/TAZ pathway-dependent manner. Interestingly, blocking exosome synthesis and secretion by GW4869 abrogated stiffness regulated motility and contractility in breast cancer cells. Reciprocally, exogenous addition of ECM stiffness-tuned exosomes orchestrated a series of changes in cell morphology, adhesion, protrusion dynamics resulting in fostered cell motility and invasion. Proteomic analysis of exosomal lysates followed by overrepresentation analysis and interactome studies revealed enrichment of cell adhesion and cell migration proteins in exosomes from stiff ECM cultures compared to that of soft ones. Quantitative proteomics of exosomes combined with genomic analysis of human breast tumor tissues (TCGA database) identified thrombospondin-1 (THBS1) as a prospective regulator of stiffness-dependent cancer invasion. Knockdown studies confirmed that the pro-invasive effects of stiffness-tuned exosomes are fuelled by exosomal THBS1. We further demonstrated that exosomal THBS1 mediates these stiffness-induced effects by engaging matrix metalloproteinase and focal adhesion kinase. Our studies establish the pivotal role of exosomal communication in ECM stiffness dependent cell migration with exosomal THBS1 as a master regulator of cancer invasion, which can be further exploited as a potential theranostic for improved breast cancer management.
Collapse
|