151
|
Parihar K, Ko SH, Bradley R, Taylor P, Ramakrishnan N, Baumgart T, Guo W, Weaver VM, Janmey PA, Radhakrishnan R. Free energy calculations for membrane morphological transformations and insights to physical biology and oncology. Methods Enzymol 2024; 701:359-386. [PMID: 39025576 PMCID: PMC11258396 DOI: 10.1016/bs.mie.2024.03.028] [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] [Indexed: 07/20/2024]
Abstract
In this chapter, we aim to bridge basic molecular and cellular principles surrounding membrane curvature generation with rewiring of cellular signals in cancer through multiscale models. We describe a general framework that integrates signaling with other cellular functions like trafficking, cell-cell and cell-matrix adhesion, and motility. The guiding question in our approach is: how does a physical change in cell membrane configuration caused by external stimuli (including those by the extracellular microenvironment) alter trafficking, signaling and subsequent cell fate? We answer this question by constructing a modeling framework based on stochastic spatial continuum models of cell membrane deformations. We apply this framework to explore the link between trafficking, signaling in the tumor microenvironment, and cell fate. At each stage, we aim to connect the results of our predictions with cellular experiments.
Collapse
Affiliation(s)
- Kshitiz Parihar
- Department of Chemical and Biomolecular Engineering, School of Engineering and Applied Science, University of Pennsylvania, Philadelphia, PA, United States
| | - Seung-Hyun Ko
- Department of Bioengineering, School of Engineering and Applied Science, University of Pennsylvania, Philadelphia, PA, United States
| | - Ryan Bradley
- Department of Chemical and Biomolecular Engineering, School of Engineering and Applied Science, University of Pennsylvania, Philadelphia, PA, United States
| | - Phillip Taylor
- Department of Chemical and Biomolecular Engineering, School of Engineering and Applied Science, University of Pennsylvania, Philadelphia, PA, United States
| | - N Ramakrishnan
- Department of Bioengineering, School of Engineering and Applied Science, University of Pennsylvania, Philadelphia, PA, United States
| | - Tobias Baumgart
- Department of Chemistry, School of Arts & Sciences, University of Pennsylvania, Philadelphia, PA, United States
| | - Wei Guo
- Department of Biology, School of Arts & Sciences, University of Pennsylvania, Philadelphia, PA, United States
| | - Valerie M Weaver
- Department of Surgery, Center for Bioengineering and Tissue Regeneration, University of California, San Francisco, San Francisco, CA, United States
| | - Paul A Janmey
- Department of Bioengineering, School of Engineering and Applied Science, University of Pennsylvania, Philadelphia, PA, United States; Department of Physiology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, United States
| | - Ravi Radhakrishnan
- Department of Chemical and Biomolecular Engineering, School of Engineering and Applied Science, University of Pennsylvania, Philadelphia, PA, United States; Department of Bioengineering, School of Engineering and Applied Science, University of Pennsylvania, Philadelphia, PA, United States.
| |
Collapse
|
152
|
Fan Y, Moser J, van Meurs M, Kiers D, Sand JMB, Leeming DJ, Pickkers P, Burgess JK, Kox M, Pillay J. Neo-epitope detection identifies extracellular matrix turnover in systemic inflammation and sepsis: an exploratory study. Crit Care 2024; 28:120. [PMID: 38609959 PMCID: PMC11010428 DOI: 10.1186/s13054-024-04904-4] [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: 01/31/2024] [Accepted: 04/06/2024] [Indexed: 04/14/2024] Open
Abstract
BACKGROUND Sepsis is associated with high morbidity and mortality, primarily due to systemic inflammation-induced tissue damage, resulting organ failure, and impaired recovery. Regulated extracellular matrix (ECM) turnover is crucial for maintaining tissue homeostasis in health and in response to disease-related changes in the tissue microenvironment. Conversely, uncontrolled turnover can contribute to tissue damage. Systemic Inflammation is implicated to play a role in the regulation of ECM turnover, but the relationship between the two is largely unclear. METHODS We performed an exploratory study in 10 healthy male volunteers who were intravenously challenged with 2 ng/kg lipopolysaccharide (LPS, derived from Escherichia coli) to induce systemic inflammation. Plasma samples were collected before (T0) and after (T 1 h, 3 h, 6 h and 24 h) the LPS challenge. Furthermore, plasma was collected from 43 patients with septic shock on day 1 of ICU admission. Circulating neo-epitopes of extracellular matrix turnover, including ECM degradation neo-epitopes of collagen type I (C1M), type III (C3M), type IV (C4Ma3), and type VI (C6M), elastin (ELP-3) and fibrin (X-FIB), as well as the ECM synthesis neo-epitopes of collagen type III (PRO-C3), collagen type IV (PRO-C4) and collagen type VI (PRO-C6) were measured by ELISA. Patient outcome data were obtained from electronic patient records. RESULTS Twenty-four hours after LPS administration, all measured ECM turnover neo-epitopes, except ELP-3, were increased compared to baseline levels. In septic shock patients, concentrations of all measured ECM neo-epitopes were higher compared to healthy controls. In addition, concentrations of C6M, ELP-3 and X-FIB were higher in patients with septic shock who ultimately did not survive (N = 7) compared to those who recovered (N = 36). CONCLUSION ECM turnover is induced in a model of systemic inflammation in healthy volunteers and was observed in patients with septic shock. Understanding interactions between systemic inflammation and ECM turnover may provide further insight into mechanisms underlying acute and persistent organ failure in sepsis.
Collapse
Affiliation(s)
- YiWen Fan
- Department of Pathology and Medical Biology, University Medical Center Groningen, University of Groningen, Groningen, The Netherlands
- Department of Critical Care, University Medical Center Groningen, University of Groningen, Hanzeplein 1, 9713 GZ, Groningen, The Netherlands
- University Medical Center Groningen, Research Institute for Asthma and COPD, University of Groningen, Groningen, The Netherlands
| | - Jill Moser
- Department of Critical Care, University Medical Center Groningen, University of Groningen, Hanzeplein 1, 9713 GZ, Groningen, The Netherlands
| | - Matijs van Meurs
- Department of Critical Care, University Medical Center Groningen, University of Groningen, Hanzeplein 1, 9713 GZ, Groningen, The Netherlands
| | - Dorien Kiers
- Department of Intensive Care Medicine, Radboud University Medical Center, Nijmegen, The Netherlands
| | | | | | - Peter Pickkers
- Department of Intensive Care Medicine, Radboud University Medical Center, Nijmegen, The Netherlands
- Radboud Centre for Infectious Diseases (RCI), Radboud University Medical Center, Nijmegen, The Netherlands
| | - Janette K Burgess
- Department of Pathology and Medical Biology, University Medical Center Groningen, University of Groningen, Groningen, The Netherlands
- University Medical Center Groningen, Research Institute for Asthma and COPD, University of Groningen, Groningen, The Netherlands
| | - Matthijs Kox
- Department of Intensive Care Medicine, Radboud University Medical Center, Nijmegen, The Netherlands
- Radboud Centre for Infectious Diseases (RCI), Radboud University Medical Center, Nijmegen, The Netherlands
| | - Janesh Pillay
- Department of Critical Care, University Medical Center Groningen, University of Groningen, Hanzeplein 1, 9713 GZ, Groningen, The Netherlands.
- University Medical Center Groningen, Research Institute for Asthma and COPD, University of Groningen, Groningen, The Netherlands.
| |
Collapse
|
153
|
Bhat AS, Chakkittukandiyil A, Muthu SK, Kotha S, Muruganandham S, Rajagopal K, Jayaram S, Kothandan R, Selvaraj D. Network-based drug repositioning of linagliptin as a potential agent for uterine fibroids targeting transforming growth factor-beta mediated fibrosis. Biochem Biophys Res Commun 2024; 703:149611. [PMID: 38354463 DOI: 10.1016/j.bbrc.2024.149611] [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: 10/23/2023] [Revised: 01/08/2024] [Accepted: 01/29/2024] [Indexed: 02/16/2024]
Abstract
Uterine fibroid is the most common non-cancerous tumor with no satisfactory options for long-term pharmacological treatment. Fibroblast activation protein-α (FAP) is one of the critical enzymes that enhances the fibrosis in uterine fibroids. Through STITCH database mining, we found that dipeptidyl peptidase-4 inhibitors (DPP4i) have the potential to inhibit the activity of FAP. Both DPP4 and FAP belong to the dipeptidyl peptidase family and share a similar catalytic domain. Hence, ligands which have a binding affinity with DPP4 could also bind with FAP. Among the DPP4i, linagliptin exhibited the highest binding affinity (Dock score = -8.562 kcal/mol) with FAP. Our study uncovered that the differences in the S2 extensive-subsite residues between DPP4 and FAP could serve as a basis for designing selective inhibitors specifically targeting FAP. Furthermore, in a dynamic environment, linagliptin was able to destabilize the dimerization interface of FAP, resulting in potential inhibition of its biological activity. True to the in-silico results, linagliptin reduced the fibrotic process in estrogen and progesterone-induced fibrosis in rat uterus. Furthermore, linagliptin reduced the gene expression of transforming growth factor-β (TGF-β), a critical factor in collagen secretion and fibrotic process. Masson trichrome staining confirmed that the anti-fibrotic effects of linagliptin were due to its ability to reduce collagen deposition in rat uterus. Altogether, our research proposes that linagliptin has the potential to be repurposed for the treatment of uterine fibroids.
Collapse
Affiliation(s)
- Anusha Shreenidhi Bhat
- Department of Pharmacology, JSS College of Pharmacy, JSS Academy of Higher Education & Research, Ooty, Nilgiris, Tamil Nadu, India
| | - Amritha Chakkittukandiyil
- Department of Pharmacology, JSS College of Pharmacy, JSS Academy of Higher Education & Research, Ooty, Nilgiris, Tamil Nadu, India
| | - Santhosh Kumar Muthu
- Department of Biochemistry, Kongunadu Arts and Science College, GN Mills, Coimbatore, Tamil Nadu, India
| | - Satvik Kotha
- Department of Pharmacology, JSS College of Pharmacy, JSS Academy of Higher Education & Research, Ooty, Nilgiris, Tamil Nadu, India
| | - Sudharsan Muruganandham
- Bioinformatics Laboratory, Department of Biotechnology, Kumaraguru College of Technology, Coimbatore, Tamil Nadu, India
| | - Kalirajan Rajagopal
- Department of Pharmaceutical Chemistry, JSS College of Pharmacy, JSS Academy of Higher Education & Research, Ooty, Nilgiris, Tamil Nadu, India
| | - Saravanan Jayaram
- Department of Pharmacology, JSS College of Pharmacy, JSS Academy of Higher Education & Research, Ooty, Nilgiris, Tamil Nadu, India
| | - Ram Kothandan
- Bioinformatics Laboratory, Department of Biotechnology, Kumaraguru College of Technology, Coimbatore, Tamil Nadu, India.
| | - Divakar Selvaraj
- Department of Pharmacology, JSS College of Pharmacy, JSS Academy of Higher Education & Research, Ooty, Nilgiris, Tamil Nadu, India.
| |
Collapse
|
154
|
Fullard N, Wordsworth J, Welsh C, Maltman V, Bascom C, Tasseff R, Isfort R, Costello L, Scanlan RL, Przyborski S, Shanley D. Cell Senescence-Independent Changes of Human Skin Fibroblasts with Age. Cells 2024; 13:659. [PMID: 38667274 PMCID: PMC11048776 DOI: 10.3390/cells13080659] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2024] [Revised: 03/15/2024] [Accepted: 03/20/2024] [Indexed: 04/28/2024] Open
Abstract
Skin ageing is defined, in part, by collagen depletion and fragmentation that leads to a loss of mechanical tension. This is currently believed to reflect, in part, the accumulation of senescent cells. We compared the expression of genes and proteins for components of the extracellular matrix (ECM) as well as their regulators and found that in vitro senescent cells produced more matrix metalloproteinases (MMPs) than proliferating cells from adult and neonatal donors. This was consistent with previous reports of senescent cells contributing to increased matrix degradation with age; however, cells from adult donors proved significantly less capable of producing new collagen than neonatal or senescent cells, and they showed significantly lower myofibroblast activation as determined by the marker α-SMA. Functionally, adult cells also showed slower migration than neonatal cells. We concluded that the increased collagen degradation of aged fibroblasts might reflect senescence, the reduced collagen production likely reflects senescence-independent processes.
Collapse
Affiliation(s)
- Nicola Fullard
- Department of Biosciences, Durham University, Durham DH1 3LE, UK
| | - James Wordsworth
- Biosciences Institute, Newcastle University, Newcastle upon Tyne NE1 7RU, UK; (J.W.); (C.W.)
| | - Ciaran Welsh
- Biosciences Institute, Newcastle University, Newcastle upon Tyne NE1 7RU, UK; (J.W.); (C.W.)
| | - Victoria Maltman
- Department of Biosciences, Durham University, Durham DH1 3LE, UK
| | | | - Ryan Tasseff
- Proctor & Gamble, Cincinnati, OH 45201, USA (R.I.)
| | | | - Lydia Costello
- Department of Biosciences, Durham University, Durham DH1 3LE, UK
| | - Rebekah-Louise Scanlan
- Biosciences Institute, Newcastle University, Newcastle upon Tyne NE1 7RU, UK; (J.W.); (C.W.)
| | | | - Daryl Shanley
- Biosciences Institute, Newcastle University, Newcastle upon Tyne NE1 7RU, UK; (J.W.); (C.W.)
| |
Collapse
|
155
|
Kopyeva I, Goldner EC, Hoye JW, Yang S, Regier MC, Vera KR, Bretherton RC, DeForest CA. Stepwise Stiffening/Softening of and Cell Recovery from Reversibly Formulated Hydrogel Double Networks. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.04.04.588191. [PMID: 38645065 PMCID: PMC11030224 DOI: 10.1101/2024.04.04.588191] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 04/23/2024]
Abstract
Biomechanical contributions of the ECM underpin cell growth and proliferation, differentiation, signal transduction, and other fate decisions. As such, biomaterials whose mechanics can be spatiotemporally altered - particularly in a reversible manner - are extremely valuable for studying these mechanobiological phenomena. Herein, we introduce a poly(ethylene glycol) (PEG)-based hydrogel model consisting of two interpenetrating step-growth networks that are independently formed via largely orthogonal bioorthogonal chemistries and sequentially degraded with distinct bacterial transpeptidases, affording reversibly tunable stiffness ranges that span healthy and diseased soft tissues (e.g., 500 Pa - 6 kPa) alongside terminal cell recovery for pooled and/or single-cell analysis in a near "biologically invisible" manner. Spatiotemporal control of gelation within the primary supporting network was achieved via mask-based and two-photon lithography; these stiffened patterned regions could be subsequently returned to the original soft state following sortase-based secondary network degradation. Using this approach, we investigated the effects of 4D-triggered network mechanical changes on human mesenchymal stem cell (hMSC) morphology and Hippo signaling, as well as Caco-2 colorectal cancer cell mechanomemory at the global transcriptome level via RNAseq. We expect this platform to be of broad utility for studying and directing mechanobiological phenomena, patterned cell fate, as well as disease resolution in softer matrices.
Collapse
Affiliation(s)
- Irina Kopyeva
- Department of Bioengineering, University of Washington, Seattle WA 98105, USA
| | - Ethan C. Goldner
- Department of Chemical Engineering, University of Washington, Seattle WA 98105, USA
| | - Jack W. Hoye
- Department of Chemical Engineering, University of Washington, Seattle WA 98105, USA
| | - Shiyu Yang
- Department of Chemical Engineering, University of Washington, Seattle WA 98105, USA
| | - Mary C. Regier
- Institute of Stem Cell & Regenerative Medicine, University of Washington, Seattle WA 98105, USA
| | - Kaitlyn R. Vera
- Department of Chemical Engineering, University of Washington, Seattle WA 98105, USA
| | - Ross C. Bretherton
- Department of Bioengineering, University of Washington, Seattle WA 98105, USA
- Institute of Stem Cell & Regenerative Medicine, University of Washington, Seattle WA 98105, USA
| | - Cole A. DeForest
- Department of Bioengineering, University of Washington, Seattle WA 98105, USA
- Department of Chemical Engineering, University of Washington, Seattle WA 98105, USA
- Institute of Stem Cell & Regenerative Medicine, University of Washington, Seattle WA 98105, USA
- Department of Chemistry, University of Washington, Seattle WA 98105, USA
- Molecular Engineering & Sciences Institute, University of Washington, Seattle WA 98105, USA
- Institute for Protein Design, University of Washington, Seattle WA 98105, USA
| |
Collapse
|
156
|
Coraggio F, Bhushan M, Roumeliotis S, Caroti F, Bevilacqua C, Prevedel R, Rapti G. Age-progressive interplay of HSP-proteostasis, ECM-cell junctions and biomechanics ensures C. elegans astroglial architecture. Nat Commun 2024; 15:2861. [PMID: 38570505 PMCID: PMC10991496 DOI: 10.1038/s41467-024-46827-2] [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: 01/08/2023] [Accepted: 03/12/2024] [Indexed: 04/05/2024] Open
Abstract
Tissue integrity is sensitive to temperature, tension, age, and is sustained throughout life by adaptive cell-autonomous or extrinsic mechanisms. Safeguarding the remarkably-complex architectures of neurons and glia ensures age-dependent integrity of functional circuits. Here, we report mechanisms sustaining the integrity of C. elegans CEPsh astrocyte-like glia. We combine large-scale genetics with manipulation of genes, cells, and their environment, quantitative imaging of cellular/ subcellular features, tissue material properties and extracellular matrix (ECM). We identify mutants with age-progressive, environment-dependent defects in glial architecture, consequent disruption of neuronal architecture, and abnormal aging. Functional loss of epithelial Hsp70/Hsc70-cochaperone BAG2 causes ECM disruption, altered tissue biomechanics, and hypersensitivity of glia to environmental temperature and mechanics. Glial-cell junctions ensure epithelia-ECM-CEPsh glia association. Modifying glial junctions or ECM mechanics safeguards glial integrity against disrupted BAG2-proteostasis. Overall, we present a finely-regulated interplay of proteostasis-ECM and cell junctions with conserved components that ensures age-progressive robustness of glial architecture.
Collapse
Affiliation(s)
- Francesca Coraggio
- Developmental Biology Unit, European Molecular Biology Laboratory, Heidelberg, Germany
| | - Mahak Bhushan
- Developmental Biology Unit, European Molecular Biology Laboratory, Heidelberg, Germany
| | - Spyridon Roumeliotis
- Developmental Biology Unit, European Molecular Biology Laboratory, Heidelberg, Germany
| | - Francesca Caroti
- Developmental Biology Unit, European Molecular Biology Laboratory, Heidelberg, Germany
| | - Carlo Bevilacqua
- Cell Biology and Biophysics Unit, European Molecular Biology Laboratory, Heidelberg, Germany
| | - Robert Prevedel
- Developmental Biology Unit, European Molecular Biology Laboratory, Heidelberg, Germany
- Cell Biology and Biophysics Unit, European Molecular Biology Laboratory, Heidelberg, Germany
- Epigenetics and Neurobiology Unit, European Molecular Biology Laboratory, Rome, Italy
- Interdisciplinary Center of Neurosciences, Heidelberg University, Heidelberg, Germany
- German Center for Lung Research (DZL), Heidelberg, Germany
| | - Georgia Rapti
- Developmental Biology Unit, European Molecular Biology Laboratory, Heidelberg, Germany.
- Epigenetics and Neurobiology Unit, European Molecular Biology Laboratory, Rome, Italy.
- Interdisciplinary Center of Neurosciences, Heidelberg University, Heidelberg, Germany.
| |
Collapse
|
157
|
Islam MT, Jang NH, Lee HJ. Natural Products as Regulators against Matrix Metalloproteinases for the Treatment of Cancer. Biomedicines 2024; 12:794. [PMID: 38672151 PMCID: PMC11048580 DOI: 10.3390/biomedicines12040794] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/29/2024] [Revised: 03/21/2024] [Accepted: 03/29/2024] [Indexed: 04/28/2024] Open
Abstract
Cancers are currently the major cause of mortality in the world. According to previous studies, matrix metalloproteinases (MMPs) have an impact on tumor cell proliferation, which could lead to the onset and progression of cancers. Therefore, regulating the expression and activity of MMPs, especially MMP-2 and MMP-9, could be a promising strategy to reduce the risk of cancers. Various studies have tried to investigate and understand the pathophysiology of cancers to suggest potent treatments. In this review, we summarize how natural products from marine organisms and plants, as regulators of MMP-2 and MMP-9 expression and enzymatic activity, can operate as potent anticancer agents.
Collapse
Affiliation(s)
- Md. Towhedul Islam
- Department of Chemistry, Faculty of Science, Mawlana Bhashani Science and Technology University, Santosh, Tangail 1902, Bangladesh
| | - Nak Han Jang
- Department of Chemistry Education, Kongju National University, Gongju 32588, Chungcheongnam-do, Republic of Korea
| | - Hyuck Jin Lee
- Department of Chemistry Education, Kongju National University, Gongju 32588, Chungcheongnam-do, Republic of Korea
| |
Collapse
|
158
|
Larsson S, Holmgren S, Jenndahl L, Ulfenborg B, Strehl R, Synnergren J, Ghosheh N. Proteome of Personalized Tissue-Engineered Veins. ACS OMEGA 2024; 9:14805-14817. [PMID: 38585136 PMCID: PMC10993322 DOI: 10.1021/acsomega.3c07098] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 09/16/2023] [Revised: 02/22/2024] [Accepted: 02/27/2024] [Indexed: 04/09/2024]
Abstract
Vascular diseases are the largest cause of death globally and impose a major global burden on healthcare. The gold standard for treating vascular diseases is the transplantation of autologous veins, if applicable. Alternative treatments still suffer from shortcomings, including low patency, lack of growth potential, the need for repeated intervention, and a substantial risk of developing infections. The use of a vascular ECM scaffold reconditioned with the patient's own cells has shown successful results in preclinical and clinical studies. In this study, we have compared the proteomes of personalized tissue-engineered veins of humans and pigs. By applying tandem mass tag (TMT) labeling LC/MS-MS, we have investigated the proteome of decellularized (DC) veins from humans and pigs and reconditioned (RC) DC veins produced through perfusion with the patient's whole blood in STEEN solution, applying the same technology as used in the preclinical studies. The results revealed high similarity between the proteomes of human and pig DC and RC veins, including the ECM texture after decellularization and reconditioning. In addition, functional enrichment analysis showed similarities in signaling pathways and biological processes involved in the immune system response. Furthermore, the classification of proteins involved in immune response activity that were detected in human and pig RC veins revealed proteins that evoke immunogenic responses, which may lead to graft rejection, thrombosis, and inflammation. However, the results from this study imply the initiation of wound healing rather than an immunogenic response, as both systems share the same processes, and no immunogenic response was reported in the preclinical and clinical studies. Finally, our study assessed the application of STEEN solution in tissue engineering and identified proteins that may be useful for the prediction of successful transplantations.
Collapse
Affiliation(s)
- Susanna Larsson
- Systems
Biology Research Center, School of Bioscience, University of Skövde, SE-541 28 Skövde, Sweden
| | - Sandra Holmgren
- VERIGRAFT, Arvid Wallgrens Backe 20, SE-413 46 Gothenburg, Sweden
| | - Lachmi Jenndahl
- VERIGRAFT, Arvid Wallgrens Backe 20, SE-413 46 Gothenburg, Sweden
| | - Benjamin Ulfenborg
- Systems
Biology Research Center, School of Bioscience, University of Skövde, SE-541 28 Skövde, Sweden
| | - Raimund Strehl
- VERIGRAFT, Arvid Wallgrens Backe 20, SE-413 46 Gothenburg, Sweden
| | - Jane Synnergren
- Systems
Biology Research Center, School of Bioscience, University of Skövde, SE-541 28 Skövde, Sweden
- Department
of Molecular and Clinical Medicine, Institute
of Medicine, Sahlgrenska Academy at University of Gothenburg, SE-413 45 Gothenburg, Sweden
| | - Nidal Ghosheh
- Systems
Biology Research Center, School of Bioscience, University of Skövde, SE-541 28 Skövde, Sweden
| |
Collapse
|
159
|
Li X, Chen W, Liu D, Chen P, Wang S, Li F, Chen Q, Lv S, Li F, Chen C, Guo S, Yuan W, Li P, Hu Z. Pathological progression of osteoarthritis: a perspective on subchondral bone. Front Med 2024; 18:237-257. [PMID: 38619691 DOI: 10.1007/s11684-024-1061-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2023] [Accepted: 01/17/2024] [Indexed: 04/16/2024]
Abstract
Osteoarthritis (OA) is a degenerative bone disease associated with aging. The rising global aging population has led to a surge in OA cases, thereby imposing a significant socioeconomic burden. Researchers have been keenly investigating the mechanisms underlying OA. Previous studies have suggested that the disease starts with synovial inflammation and hyperplasia, advancing toward cartilage degradation. Ultimately, subchondral-bone collapse, sclerosis, and osteophyte formation occur. This progression is deemed as "top to bottom." However, recent research is challenging this perspective by indicating that initial changes occur in subchondral bone, precipitating cartilage breakdown. In this review, we elucidate the epidemiology of OA and present an in-depth overview of the subchondral bone's physiological state, functions, and the varied pathological shifts during OA progression. We also introduce the role of multifunctional signal pathways (including osteoprotegerin (OPG)/receptor activator of nuclear factor-kappa B ligand (RANKL)/receptor activator of nuclear factor-kappa B (RANK), and chemokine (CXC motif) ligand 12 (CXCL12)/CXC motif chemokine receptor 4 (CXCR4)) in the pathology of subchondral bone and their role in the "bottom-up" progression of OA. Using vivid pattern maps and clinical images, this review highlights the crucial role of subchondral bone in driving OA progression, illuminating its interplay with the condition.
Collapse
Affiliation(s)
- Xuefei Li
- Longhua Hospital Affiliated to Shanghai University of Traditional Chinese Medicine, Shanghai, 200032, China
| | - Wenhua Chen
- Research and Development Center of Chinese Medicine Resources and Biotechnology, Shanghai University of Traditional Chinese Medicine, Shanghai, 201203, China
| | - Dan Liu
- Longhua Hospital Affiliated to Shanghai University of Traditional Chinese Medicine, Shanghai, 200032, China
| | - Pinghua Chen
- Longhua Hospital Affiliated to Shanghai University of Traditional Chinese Medicine, Shanghai, 200032, China
| | - Shiyun Wang
- Longhua Hospital Affiliated to Shanghai University of Traditional Chinese Medicine, Shanghai, 200032, China
| | - Fangfang Li
- Longhua Hospital Affiliated to Shanghai University of Traditional Chinese Medicine, Shanghai, 200032, China
| | - Qian Chen
- Longhua Hospital Affiliated to Shanghai University of Traditional Chinese Medicine, Shanghai, 200032, China
| | - Shunyi Lv
- Longhua Hospital Affiliated to Shanghai University of Traditional Chinese Medicine, Shanghai, 200032, China
| | - Fangyu Li
- Longhua Hospital Affiliated to Shanghai University of Traditional Chinese Medicine, Shanghai, 200032, China
| | - Chen Chen
- Longhua Hospital Affiliated to Shanghai University of Traditional Chinese Medicine, Shanghai, 200032, China
| | - Suxia Guo
- Longhua Hospital Affiliated to Shanghai University of Traditional Chinese Medicine, Shanghai, 200032, China
| | - Weina Yuan
- Longhua Hospital Affiliated to Shanghai University of Traditional Chinese Medicine, Shanghai, 200032, China
| | - Pan Li
- Longhua Hospital Affiliated to Shanghai University of Traditional Chinese Medicine, Shanghai, 200032, China
| | - Zhijun Hu
- Longhua Hospital Affiliated to Shanghai University of Traditional Chinese Medicine, Shanghai, 200032, China.
| |
Collapse
|
160
|
Zhao JY, Zhou LJ, Ma KL, Hao R, Li M. MHO or MUO? White adipose tissue remodeling. Obes Rev 2024; 25:e13691. [PMID: 38186200 DOI: 10.1111/obr.13691] [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: 05/05/2023] [Revised: 11/14/2023] [Accepted: 11/19/2023] [Indexed: 01/09/2024]
Abstract
In this review, we delve into the intricate relationship between white adipose tissue (WAT) remodeling and metabolic aspects in obesity, with a specific focus on individuals with metabolically healthy obesity (MHO) and metabolically unhealthy obesity (MUO). WAT is a highly heterogeneous, plastic, and dynamically secreting endocrine and immune organ. WAT remodeling plays a crucial role in metabolic health, involving expansion mode, microenvironment, phenotype, and distribution. In individuals with MHO, WAT remodeling is beneficial, reducing ectopic fat deposition and insulin resistance (IR) through mechanisms like increased adipocyte hyperplasia, anti-inflammatory microenvironment, appropriate extracellular matrix (ECM) remodeling, appropriate vascularization, enhanced WAT browning, and subcutaneous adipose tissue (SWAT) deposition. Conversely, for those with MUO, WAT remodeling leads to ectopic fat deposition and IR, causing metabolic dysregulation. This process involves adipocyte hypertrophy, disrupted vascularization, heightened pro-inflammatory microenvironment, enhanced brown adipose tissue (BAT) whitening, and accumulation of visceral adipose tissue (VWAT) deposition. The review underscores the pivotal importance of intervening in WAT remodeling to hinder the transition from MHO to MUO. This insight is valuable for tailoring personalized and effective management strategies for patients with obesity in clinical practice.
Collapse
Affiliation(s)
- Jing Yi Zhao
- Research Laboratory of Molecular Biology, Guang'anmen Hospital, China Academy of Chinese Medical Sciences, Beijing, China
| | - Li Juan Zhou
- Research Laboratory of Molecular Biology, Guang'anmen Hospital, China Academy of Chinese Medical Sciences, Beijing, China
| | - Kai Le Ma
- Research Laboratory of Molecular Biology, Guang'anmen Hospital, China Academy of Chinese Medical Sciences, Beijing, China
| | - Rui Hao
- Research Laboratory of Molecular Biology, Guang'anmen Hospital, China Academy of Chinese Medical Sciences, Beijing, China
| | - Min Li
- Research Laboratory of Molecular Biology, Guang'anmen Hospital, China Academy of Chinese Medical Sciences, Beijing, China
| |
Collapse
|
161
|
Giunashvili N, Thomas JM, Schvarcz CA, Viana PHL, Aloss K, Bokhari SMZ, Koós Z, Bócsi D, Major E, Balogh A, Benyó Z, Hamar P. Enhancing therapeutic efficacy in triple-negative breast cancer and melanoma: synergistic effects of modulated electro-hyperthermia (mEHT) with NSAIDs especially COX-2 inhibition in in vivo models. Mol Oncol 2024; 18:1012-1030. [PMID: 38217262 PMCID: PMC10994232 DOI: 10.1002/1878-0261.13585] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2023] [Revised: 12/18/2023] [Accepted: 01/02/2024] [Indexed: 01/15/2024] Open
Abstract
Triple-negative breast cancer (TNBC) is a leading cause of cancer mortality and lacks modern therapy options. Modulated electro-hyperthermia (mEHT) is an adjuvant therapy with demonstrated clinical efficacy for the treatment of various cancer types. In this study, we report that mEHT monotherapy stimulated interleukin-1 beta (IL-1β) and interleukin-6 (IL-6) expression, and consequently cyclooxygenase 2 (COX-2), which may favor a cancer-promoting tumor microenvironment. Thus, we combined mEHT with nonsteroid anti-inflammatory drugs (NSAIDs): a nonselective aspirin, or the selective COX-2 inhibitor SC236, in vivo. We demonstrate that NSAIDs synergistically increased the effect of mEHT in the 4T1 TNBC model. Moreover, the strongest tumor destruction ratio was observed in the combination SC236 + mEHT groups. Tumor damage was accompanied by a significant increase in cleaved caspase-3, suggesting that apoptosis played an important role. IL-1β and COX-2 expression were significantly reduced by the combination therapies. In addition, a custom-made nanostring panel demonstrated significant upregulation of genes participating in the formation of the extracellular matrix. Similarly, in the B16F10 melanoma model, mEHT and aspirin synergistically reduced the number of melanoma nodules in the lungs. In conclusion, mEHT combined with a selective COX-2 inhibitor may offer a new therapeutic option in TNBC.
Collapse
Grants
- STIA-OTKA-2022 Semmelweis Science and Innovation Fund
- OTKA_ANN 110810 National Research, Development, and Innovation Office
- OTKA_SNN 114619 National Research, Development, and Innovation Office
- ÚNKP-23-3-II-SE-45 National Research, Development, and Innovation Office
- ÚNKP-23-4-I-SE-22 National Research, Development, and Innovation Office
- OTKA_K 145998 National Research, Development, and Innovation Office
- Tempus Foundation
- EFOP-3.6.3-VEKOP-16-2017-00009 Semmelweis Excellence 250+ Scholarship
Collapse
Affiliation(s)
- Nino Giunashvili
- Institute of Translational Medicine, Semmelweis UniversityBudapestHungary
| | | | - Csaba András Schvarcz
- Institute of Translational Medicine, Semmelweis UniversityBudapestHungary
- HUN‐REN‐SU Cerebrovascular and Neurocognitive Diseases Research GroupBudapestHungary
| | | | - Kenan Aloss
- Institute of Translational Medicine, Semmelweis UniversityBudapestHungary
| | | | - Zoltán Koós
- Institute of Translational Medicine, Semmelweis UniversityBudapestHungary
| | - Dániel Bócsi
- Institute of Translational Medicine, Semmelweis UniversityBudapestHungary
| | - Enikő Major
- Institute of Translational Medicine, Semmelweis UniversityBudapestHungary
- HUN‐REN‐SU Cerebrovascular and Neurocognitive Diseases Research GroupBudapestHungary
| | - Andrea Balogh
- Institute of Translational Medicine, Semmelweis UniversityBudapestHungary
| | - Zoltán Benyó
- Institute of Translational Medicine, Semmelweis UniversityBudapestHungary
- HUN‐REN‐SU Cerebrovascular and Neurocognitive Diseases Research GroupBudapestHungary
| | - Péter Hamar
- Institute of Translational Medicine, Semmelweis UniversityBudapestHungary
| |
Collapse
|
162
|
Stock C. pH-regulated single cell migration. Pflugers Arch 2024; 476:639-658. [PMID: 38214759 PMCID: PMC11006768 DOI: 10.1007/s00424-024-02907-2] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2023] [Revised: 12/21/2023] [Accepted: 01/02/2024] [Indexed: 01/13/2024]
Abstract
Over the last two decades, extra- and intracellular pH have emerged as fundamental regulators of cell motility. Fundamental physiological and pathological processes relying on appropriate cell migration, such as embryonic development, wound healing, and a proper immune defense on the one hand, and autoimmune diseases, metastatic cancer, and the progression of certain parasitic diseases on the other, depend on surrounding pH. In addition, migrating single cells create their own localized pH nanodomains at their surface and in the cytosol. By this means, the migrating cells locally modulate their adhesion to, and the re-arrangement and digestion of, the extracellular matrix. At the same time, the cytosolic nanodomains tune cytoskeletal dynamics along the direction of movement resulting in concerted lamellipodia protrusion and rear end retraction. Extracellular pH gradients as found in wounds, inflamed tissues, or the periphery of tumors stimulate directed cell migration, and long-term exposure to acidic conditions can engender a more migratory and invasive phenotype persisting for hours up to several generations of cells after they have left the acidic milieu. In the present review, the different variants of pH-dependent single cell migration are described. The underlying pH-dependent molecular mechanisms such as conformational changes of adhesion molecules, matrix protease activity, actin (de-)polymerization, and signaling events are explained, and molecular pH sensors stimulated by H+ signaling are presented.
Collapse
Affiliation(s)
- Christian Stock
- Department of Gastroenterology, Hepatology, Infectiology & Endocrinology, Hannover Medical School, Carl-Neuberg-Str. 1, 30625, Hannover, Germany.
| |
Collapse
|
163
|
Majumder N, Seit S, Bhabesh NS, Ghosh S. An Advanced Bioconjugation Strategy for Covalent Tethering of TGFβ3 with Silk Fibroin Matrices and its Implications in the Chondrogenesis Profile of Human BMSCs and Human Chondrocytes: A Paradigm Shift in Cartilage Tissue Engineering. Adv Healthc Mater 2024; 13:e2303513. [PMID: 38291832 DOI: 10.1002/adhm.202303513] [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: 10/13/2023] [Revised: 01/25/2024] [Indexed: 02/01/2024]
Abstract
The transforming growth factor-β class of cytokines plays a significant role in articular cartilage formation from mesenchymal condensation to chondrogenic differentiation. However, their exogenous addition to the chondrogenic media makes the protocol expensive. It reduces the bioavailability of the cytokine to the cells owing to their burst release. The present study demonstrates an advanced bioconjugation strategy to conjugate transforming growth factor-β3 (TGFβ3) with silk fibroin matrix covalently via a cyanuric chloride coupling reaction. The tethering and change in secondary conformation are confirmed using various spectroscopic analyses. To assess the functionality of the chemically modified silk matrix, human bone marrow-derived mesenchymal stem cells (hBMSCs) and chondrocytes are cultured for 28 days in a chondrogenic differentiation medium. Gene expression and histological analysis reveal enhanced expression of chondrogenic markers with intense Safranin-O and Alcian Blue staining in TGFβ3 conjugated silk matrices than where TGFβ3 is exogenously added to the media for both hBMSCs and chondrocytes. Therefore, this study successfully recapitulates the native niche of TGFβ3 and the role of the silk as a growth factor stabilizer. When cultured over TGFβ3 conjugated silk matrices, hBMSCs display increased proteoglycan secretion and maximum chondrogenic trait with attenuation of chondrocyte hypertrophy over human chondrocytes.
Collapse
Affiliation(s)
- Nilotpal Majumder
- Department of Textile and Fibre Engineering, Indian Institute of Technology Delhi, New Delhi, 110016, India
| | - Sinchan Seit
- Department of Textile and Fibre Engineering, Indian Institute of Technology Delhi, New Delhi, 110016, India
| | - Neel Sarovar Bhabesh
- International Centre for Genetic Engineering and Biotechnology (ICGEB), Transcription Regulation group, New Delhi, 110067, India
| | - Sourabh Ghosh
- Department of Textile and Fibre Engineering, Indian Institute of Technology Delhi, New Delhi, 110016, India
| |
Collapse
|
164
|
Vlashi R, Zhang X, Li H, Chen G. Potential therapeutic strategies for osteoarthritis via CRISPR/Cas9 mediated gene editing. Rev Endocr Metab Disord 2024; 25:339-367. [PMID: 38055160 DOI: 10.1007/s11154-023-09860-y] [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] [Accepted: 11/28/2023] [Indexed: 12/07/2023]
Abstract
Osteoarthritis (OA) is an incapacitating and one of the most common physically degenerative conditions with an assorted etiology and a highly complicated molecular mechanism that to date lacks an efficient treatment. The capacity to design biological networks and accurately modify existing genomic sites holds an apt potential for applications across medical and biotechnological sciences. One of these highly specific genomes editing technologies is the CRISPR/Cas9 mechanism, referred to as the clustered regularly interspaced short palindromic repeats, which is a defense mechanism constituted by CRISPR associated protein 9 (Cas9) directed by small non-coding RNAs (sncRNA) that bind to target DNA through Watson-Crick base pairing rules where subsequent repair of the target DNA is initiated. Up-to-date research has established the effectiveness of the CRISPR/Cas9 mechanism in targeting the genetic and epigenetic alterations in OA by suppressing or deleting gene expressions and eventually distributing distinctive anti-arthritic properties in both in vitro and in vivo osteoarthritic models. This review aims to epitomize the role of this high-throughput and multiplexed gene editing method as an analogous therapeutic strategy that could greatly facilitate the clinical development of OA-related treatments since it's reportedly an easy, minimally invasive technique, and a comparatively less painful method for osteoarthritic patients.
Collapse
Affiliation(s)
- Rexhina Vlashi
- College of Life Science and Medicine, Zhejiang Provincial Key Laboratory of Silkworm Bioreactor and Biomedicine, Zhejiang Sci-Tech University, Hangzhou, 310018, China
| | - Xingen Zhang
- Department of Orthopedics, Jiaxing Key Laboratory for Minimally Invasive Surgery in Orthopaedics & Skeletal Regenerative Medicine, Zhejiang Rongjun Hospital, Jiaxing, 314001, China
| | - Haibo Li
- The Central Laboratory of Birth Defects Prevention and Control, Ningbo Women and Children's Hospital, Ningbo, China.
- Ningbo Key Laboratory for the Prevention and Treatment of Embryogenic Diseases, Ningbo Women and Children's Hospital, Ningbo, China.
| | - Guiqian Chen
- College of Life Science and Medicine, Zhejiang Provincial Key Laboratory of Silkworm Bioreactor and Biomedicine, Zhejiang Sci-Tech University, Hangzhou, 310018, China.
| |
Collapse
|
165
|
Djalali-Cuevas A, Rettel M, Stein F, Savitski M, Kearns S, Kelly J, Biggs M, Skoufos I, Tzora A, Prassinos N, Diakakis N, Zeugolis DI. Macromolecular crowding in human tenocyte and skin fibroblast cultures: A comparative analysis. Mater Today Bio 2024; 25:100977. [PMID: 38322661 PMCID: PMC10846491 DOI: 10.1016/j.mtbio.2024.100977] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2023] [Revised: 12/22/2023] [Accepted: 01/24/2024] [Indexed: 02/08/2024] Open
Abstract
Although human tenocytes and dermal fibroblasts have shown promise in tendon engineering, no tissue engineered medicine has been developed due to the prolonged ex vivo time required to develop an implantable device. Considering that macromolecular crowding has the potential to substantially accelerate the development of functional tissue facsimiles, herein we compared human tenocyte and dermal fibroblast behaviour under standard and macromolecular crowding conditions to inform future studies in tendon engineering. Basic cell function analysis made apparent the innocuousness of macromolecular crowding for both cell types. Gene expression analysis of the without macromolecular crowding groups revealed expression of tendon related molecules in human dermal fibroblasts and tenocytes. Protein electrophoresis and immunocytochemistry analyses showed significantly increased and similar deposition of collagen fibres by macromolecular crowding in the two cell types. Proteomics analysis demonstrated great similarities between human tenocyte and dermal fibroblast cultures, as well as the induction of haemostatic, anti-microbial and tissue-protective proteins by macromolecular crowding in both cell populations. Collectively, these data rationalise the use of either human dermal fibroblasts or tenocytes in combination with macromolecular crowding in tendon engineering.
Collapse
Affiliation(s)
- Adrian Djalali-Cuevas
- Laboratory of Animal Science, Nutrition and Biotechnology, School of Agriculture, University of Ioannina, Arta, Greece
- School of Veterinary Medicine, Aristotle University of Thessaloniki, Thessaloniki, Greece
- Regenerative, Modular & Developmental Engineering Laboratory (REMODEL), Charles Institute of Dermatology, Conway Institute of Biomolecular & Biomedical Research and School of Mechanical & Materials Engineering, University College Dublin (UCD), Dublin, Ireland
| | - Mandy Rettel
- Proteomics Core Facility, European Molecular Biology Laboratory (EMBL), Heidelberg, Germany
| | - Frank Stein
- Proteomics Core Facility, European Molecular Biology Laboratory (EMBL), Heidelberg, Germany
| | - Mikhail Savitski
- Proteomics Core Facility, European Molecular Biology Laboratory (EMBL), Heidelberg, Germany
| | | | - Jack Kelly
- Galway University Hospital, Galway, Ireland
| | - Manus Biggs
- Science Foundation Ireland (SFI) Centre for Research in Medical Devices (CÚRAM), Biomedical Sciences Building, University of Galway, Galway, Ireland
| | - Ioannis Skoufos
- Laboratory of Animal Science, Nutrition and Biotechnology, School of Agriculture, University of Ioannina, Arta, Greece
| | - Athina Tzora
- Laboratory of Animal Science, Nutrition and Biotechnology, School of Agriculture, University of Ioannina, Arta, Greece
| | - Nikitas Prassinos
- School of Veterinary Medicine, Aristotle University of Thessaloniki, Thessaloniki, Greece
| | - Nikolaos Diakakis
- School of Veterinary Medicine, Aristotle University of Thessaloniki, Thessaloniki, Greece
| | - Dimitrios I. Zeugolis
- Regenerative, Modular & Developmental Engineering Laboratory (REMODEL), Charles Institute of Dermatology, Conway Institute of Biomolecular & Biomedical Research and School of Mechanical & Materials Engineering, University College Dublin (UCD), Dublin, Ireland
| |
Collapse
|
166
|
Fu Y, Zhou Y, Wang K, Li Z, Kong W. Extracellular Matrix Interactome in Modulating Vascular Homeostasis and Remodeling. Circ Res 2024; 134:931-949. [PMID: 38547250 DOI: 10.1161/circresaha.123.324055] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 04/02/2024]
Abstract
The ECM (extracellular matrix) is a major component of the vascular microenvironment that modulates vascular homeostasis. ECM proteins include collagens, elastin, noncollagen glycoproteins, and proteoglycans/glycosaminoglycans. ECM proteins form complex matrix structures, such as the basal lamina and collagen and elastin fibers, through direct interactions or lysyl oxidase-mediated cross-linking. Moreover, ECM proteins directly interact with cell surface receptors or extracellular secreted molecules, exerting matricellular and matricrine modulation, respectively. In addition, extracellular proteases degrade or cleave matrix proteins, thereby contributing to ECM turnover. These interactions constitute the ECM interactome network, which is essential for maintaining vascular homeostasis and preventing pathological vascular remodeling. The current review mainly focuses on endogenous matrix proteins in blood vessels and discusses the interaction of these matrix proteins with other ECM proteins, cell surface receptors, cytokines, complement and coagulation factors, and their potential roles in maintaining vascular homeostasis and preventing pathological remodeling.
Collapse
Affiliation(s)
- Yi Fu
- Department of Physiology and Pathophysiology (Y.F., K.W., Z.L., W.K.), School of Basic Medical Sciences, State Key Laboratory of Vascular Homeostasis and Remodeling, Peking University, Beijing, China
| | - Yuan Zhou
- Department of Biomedical Informatics (Y.Z.), School of Basic Medical Sciences, State Key Laboratory of Vascular Homeostasis and Remodeling, Peking University, Beijing, China
| | - Kai Wang
- Department of Physiology and Pathophysiology (Y.F., K.W., Z.L., W.K.), School of Basic Medical Sciences, State Key Laboratory of Vascular Homeostasis and Remodeling, Peking University, Beijing, China
| | - Zhuofan Li
- Department of Physiology and Pathophysiology (Y.F., K.W., Z.L., W.K.), School of Basic Medical Sciences, State Key Laboratory of Vascular Homeostasis and Remodeling, Peking University, Beijing, China
| | - Wei Kong
- Department of Physiology and Pathophysiology (Y.F., K.W., Z.L., W.K.), School of Basic Medical Sciences, State Key Laboratory of Vascular Homeostasis and Remodeling, Peking University, Beijing, China
| |
Collapse
|
167
|
Price MJ, Nguyen AD, Haines C, Baëta CD, Byemerwa J, Murkajee D, Artham S, Kumar V, Lavau C, Wardell S, Varghese S, Goodwin CR. UDP-6-glucose dehydrogenase in hormonally responsive breast cancers. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.03.20.585919. [PMID: 38562874 PMCID: PMC10983948 DOI: 10.1101/2024.03.20.585919] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 04/04/2024]
Abstract
Survival for metastatic breast cancer is low and thus, continued efforts to treat and prevent metastatic progression are critical. Estrogen is shown to promote aggressive phenotypes in multiple cancer models irrespective of estrogen receptor (ER) status. Similarly, UDP-Glucose 6-dehydrogenase (UGDH) a ubiquitously expressed enzyme involved in extracellular matrix precursors, as well as hormone processing increases migratory and invasive properties in cancer models. While the role of UGDH in cellular migration is defined, how it intersects with and impacts hormone signaling pathways associated with tumor progression in metastatic breast cancer has not been explored. Here we demonstrate that UGDH knockdown blunts estrogen-induced tumorigenic phenotypes (migration and colony formation) in ER+ and ER- breast cancer in vitro. Knockdown of UGDH also inhibits extravasation of ER- breast cancer ex vivo, primary tumor growth and animal survival in vivo in both ER+ and ER- breast cancer. We also use single cell RNA-sequencing to demonstrate that our findings translate to a human breast cancer clinical specimen. Our findings support the role of estrogen and UGDH in breast cancer progression provide a foundation for future studies to evaluate the role of UGDH in therapeutic resistance to improve outcomes and survival for breast cancer patients.
Collapse
Affiliation(s)
- Meghan J Price
- Department of Neurosurgery, Duke University Medical Center, University School of Medicine, Durham, NC, USA
- Department of Medicine, John Hopkins Hospital, 1800 Orleans St, Baltimore, MD 21287, USA
| | - Annee D Nguyen
- Department of Neurosurgery, Duke University Medical Center, University School of Medicine, Durham, NC, USA
| | - Corinne Haines
- Department of Molecular Genetics, Ohio State University, 1060 Carmack Road, Columbus, OH 43210, USA
| | - César D Baëta
- Department of Neurosurgery, Duke University Medical Center, University School of Medicine, Durham, NC, USA
- Center for Population Health Sciences, Stanford University, 1701 Page Mill Road, Palo Alto, CA 94304, USA
| | - Jovita Byemerwa
- Department of Pharmacology and Cancer Biology, Duke University Medical Center, University School of Medicine, Durham, NC, USA
| | - Debarati Murkajee
- Department of Pharmacology and Cancer Biology, Duke University Medical Center, University School of Medicine, Durham, NC, USA
| | - Sandeep Artham
- Department of Pharmacology and Cancer Biology, Duke University Medical Center, University School of Medicine, Durham, NC, USA
| | - Vardhman Kumar
- Department of Biomedical Engineering, Duke University Medical Center, Durham, NC, USA
| | - Catherine Lavau
- Department of Neurosurgery, Duke University Medical Center, University School of Medicine, Durham, NC, USA
| | - Suzanne Wardell
- Department of Pharmacology and Cancer Biology, Duke University Medical Center, University School of Medicine, Durham, NC, USA
| | - Shyni Varghese
- Department of Biomedical Engineering, Duke University Medical Center, Durham, NC, USA
- Department of Orthopedic Surgery, Duke University Medical Center, Durham, NC, USA
| | - C Rory Goodwin
- Department of Neurosurgery, Duke University Medical Center, University School of Medicine, Durham, NC, USA
| |
Collapse
|
168
|
Sharip A, Rakhimova S, Molkenov A, Ashenova A, Kozhamkulov U, Akhmetollayev I, Zinovyev A, Zhukov Y, Omarov M, Tuleutaev M, Rakhmetova V, Terwilliger JD, Lee JH, Zhumadilov Z, Akilzhanova A, Kairov U. Transcriptome profiling and analysis of patients with esophageal squamous cell carcinoma from Kazakhstan. Front Genet 2024; 15:1249751. [PMID: 38562378 PMCID: PMC10982404 DOI: 10.3389/fgene.2024.1249751] [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: 06/29/2023] [Accepted: 03/06/2024] [Indexed: 04/04/2024] Open
Abstract
Esophageal squamous cell carcinoma (ESCC) is the predominant subtype of esophageal cancer in Central Asia, often diagnosed at advanced stages. Understanding population-specific patterns of ESCC is crucial for tailored treatments. This study aimed to unravel ESCC's genetic basis in Kazakhstani patients and identify potential biomarkers for early diagnosis and targeted therapies. ESCC patients from Kazakhstan were studied. We analyzed histological subtypes and conducted in-depth transcriptome sequencing. Differential gene expression analysis was performed, and significantly dysregulated pathways were identified using KEGG pathway analysis (p-value < 0.05). Protein-protein interaction networks were constructed to elucidate key modules and their functions. Among Kazakhstani patients, ESCC with moderate dysplasia was the most prevalent subtype. We identified 42 significantly upregulated and two significantly downregulated KEGG pathways, highlighting molecular mechanisms driving ESCC pathogenesis. Immune-related pathways, such as viral protein interaction with cytokines, rheumatoid arthritis, and oxidative phosphorylation, were elevated, suggesting immune system involvement. Conversely, downregulated pathways were associated with extracellular matrix degradation, crucial in cancer invasion and metastasis. Protein-protein interaction network analysis revealed four distinct modules with specific functions, implicating pathways in esophageal cancer development. High-throughput transcriptome sequencing elucidated critical molecular pathways underlying esophageal carcinogenesis in Kazakhstani patients. Insights into dysregulated pathways offer potential for early diagnosis and precision treatment strategies for ESCC. Understanding population-specific patterns is essential for personalized approaches to ESCC management.
Collapse
Affiliation(s)
- Aigul Sharip
- Center for Life Sciences, National Laboratory Astana, Nazarbayev University, Astana, Kazakhstan
| | - Saule Rakhimova
- Center for Life Sciences, National Laboratory Astana, Nazarbayev University, Astana, Kazakhstan
| | - Askhat Molkenov
- Center for Life Sciences, National Laboratory Astana, Nazarbayev University, Astana, Kazakhstan
| | - Ainur Ashenova
- Center for Life Sciences, National Laboratory Astana, Nazarbayev University, Astana, Kazakhstan
| | - Ulan Kozhamkulov
- Center for Life Sciences, National Laboratory Astana, Nazarbayev University, Astana, Kazakhstan
| | | | | | - Yuri Zhukov
- Multidisciplinary Medical Center, Astana, Kazakhstan
| | - Marat Omarov
- Multidisciplinary Medical Center, Astana, Kazakhstan
| | | | - Venera Rakhmetova
- Department of Internal Diseases, Astana Medical University, Astana, Kazakhstan
| | - Joseph D. Terwilliger
- Sergiеvsky Center, Columbia University, New York, NY, United States
- Division of Medical Genetics, New York State Psychiatric Institute, New York, NY, United States
- Department of Psychiatry and Department of Genetics and Development, Columbia University, New York, NY, United States
| | - Joseph H. Lee
- Sergiеvsky Center, Columbia University, New York, NY, United States
- Departments of Epidemiology and Neurology, Columbia University, New York, NY, United States
| | - Zhaxybay Zhumadilov
- Center for Life Sciences, National Laboratory Astana, Nazarbayev University, Astana, Kazakhstan
- School of Medicine, Nazarbayev University, Astana, Kazakhstan
| | - Ainur Akilzhanova
- Center for Life Sciences, National Laboratory Astana, Nazarbayev University, Astana, Kazakhstan
| | - Ulykbek Kairov
- Center for Life Sciences, National Laboratory Astana, Nazarbayev University, Astana, Kazakhstan
| |
Collapse
|
169
|
Meizlish ML, Kimura Y, Pope SD, Matta R, Kim C, Philip NH, Meyaard L, Gonzalez A, Medzhitov R. Mechanosensing regulates tissue repair program in macrophages. SCIENCE ADVANCES 2024; 10:eadk6906. [PMID: 38478620 PMCID: PMC10936955 DOI: 10.1126/sciadv.adk6906] [Citation(s) in RCA: 16] [Impact Index Per Article: 16.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 09/05/2023] [Accepted: 01/29/2024] [Indexed: 03/17/2024]
Abstract
Tissue-resident macrophages play important roles in tissue homeostasis and repair. However, how macrophages monitor and maintain tissue integrity is not well understood. The extracellular matrix (ECM) is a key structural and organizational component of all tissues. Here, we find that macrophages sense the mechanical properties of the ECM to regulate a specific tissue repair program. We show that macrophage mechanosensing is mediated by cytoskeletal remodeling and can be performed in three-dimensional environments through a noncanonical, integrin-independent mechanism analogous to amoeboid migration. We find that these cytoskeletal dynamics also integrate biochemical signaling by colony-stimulating factor 1 and ultimately regulate chromatin accessibility to control the mechanosensitive gene expression program. This study identifies an "amoeboid" mode of ECM mechanosensing through which macrophages may regulate tissue repair and fibrosis.
Collapse
Affiliation(s)
- Matthew L. Meizlish
- Department of Immunobiology, Yale University School of Medicine, New Haven, CT, USA
- Department of Medicine, Massachusetts General Hospital, Boston, MA, USA
| | - Yoshitaka Kimura
- Department of Immunobiology, Yale University School of Medicine, New Haven, CT, USA
| | - Scott D. Pope
- Department of Immunobiology, Yale University School of Medicine, New Haven, CT, USA
| | - Rita Matta
- Department of Biomedical Engineering, Yale University, New Haven, CT, USA
| | - Catherine Kim
- Department of Biomedical Engineering, Yale University, New Haven, CT, USA
| | - Naomi H. Philip
- Department of Immunobiology, Yale University School of Medicine, New Haven, CT, USA
| | - Linde Meyaard
- Center for Translational Immunology, University Medical Center Utrecht, Utrecht University, Utrecht, Netherlands
- Oncode Institute, Utrecht, Netherlands
| | - Anjelica Gonzalez
- Department of Biomedical Engineering, Yale University, New Haven, CT, USA
| | - Ruslan Medzhitov
- Department of Immunobiology, Yale University School of Medicine, New Haven, CT, USA
- Howard Hughes Medical Institute, Yale University School of Medicine, New Haven, CT, USA
| |
Collapse
|
170
|
Shi Z, Liu Z, Wei Y, Zhang R, Deng Y, Li D. The role of dermal fibroblasts in autoimmune skin diseases. Front Immunol 2024; 15:1379490. [PMID: 38545113 PMCID: PMC10965632 DOI: 10.3389/fimmu.2024.1379490] [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: 01/31/2024] [Accepted: 03/01/2024] [Indexed: 04/18/2024] Open
Abstract
Fibroblasts are an important subset of mesenchymal cells in maintaining skin homeostasis and resisting harmful stimuli. Meanwhile, fibroblasts modulate immune cell function by secreting cytokines, thereby implicating their involvement in various dermatological conditions such as psoriasis, vitiligo, and atopic dermatitis. Recently, variations in the subtypes of fibroblasts and their expression profiles have been identified in these prevalent autoimmune skin diseases, implying that fibroblasts may exhibit distinct functionalities across different diseases. In this review, from the perspective of their fundamental functions and remarkable heterogeneity, we have comprehensively collected evidence on the role of fibroblasts and their distinct subpopulations in psoriasis, vitiligo, atopic dermatitis, and scleroderma. Importantly, these findings hold promise for guiding future research directions and identifying novel therapeutic targets for treating these diseases.
Collapse
Affiliation(s)
| | | | | | | | | | - Dong Li
- Department of Dermatology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| |
Collapse
|
171
|
Altieri A, Marshall CL, Ramotar P, Lloyd D, Hemshekhar M, Spicer V, van der Does AM, Mookherjee N. Human Host Defense Peptide LL-37 Suppresses TNFα-Mediated Matrix Metalloproteinases MMP9 and MMP13 in Human Bronchial Epithelial Cells. J Innate Immun 2024; 16:203-215. [PMID: 38471488 PMCID: PMC10997319 DOI: 10.1159/000537775] [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: 09/29/2023] [Accepted: 02/09/2024] [Indexed: 03/14/2024] Open
Abstract
INTRODUCTION TNFα-inducible matrix metalloproteinases play a critical role in the process of airway remodeling in respiratory inflammatory disease including asthma. The cationic host defense peptide LL-37 is elevated in the lungs during airway inflammation. However, the impact of LL-37 on TNFα-driven processes is not well understood. Here, we examined the effect of LL-37 on TNFα-mediated responses in human bronchial epithelial cells (HBECs). METHODS We used a slow off-rate modified aptamer-based proteomics approach to define the HBEC proteome altered in response to TNFα. Abundance of selected protein candidates and signaling intermediates was examined using immunoassays, ELISA and Western blots, and mRNA abundance was examined by qRT-PCR. RESULTS Proteomics analysis revealed that 124 proteins were significantly altered, 12 proteins were enhanced by ≥2-fold compared to unstimulated cells, in response to TNFα. MMP9 was the topmost increased protein in response to TNFα, enhanced by ∼10-fold, and MMP13 was increased by ∼3-fold, compared to unstimulated cells. Furthermore, we demonstrated that LL-37 significantly suppressed TNFα-mediated MMP9 and MMP13 in HBEC. Mechanistic data revealed that TNFα-mediated MMP9 and MMP13 production is controlled by SRC kinase and that LL-37 enhances related upstream negative regulators, namely, phospho-AKT (T308) and TNFα-mediated TNFAIP3 or A20. CONCLUSIONS The findings of this study suggest that LL-37 may play a role in intervening in the process of airway remodeling in chronic inflammatory respiratory disease such as asthma.
Collapse
Affiliation(s)
- Anthony Altieri
- Department of Immunology, University of Manitoba, Winnipeg, MB, Canada
- Department of Immunology, University of Toronto, Toronto, ON, Canada
| | | | - Padmanie Ramotar
- Department of Immunology, University of Manitoba, Winnipeg, MB, Canada
| | - Dylan Lloyd
- Manitoba Centre for Proteomics and Systems Biology, Department of Internal Medicine, University of Manitoba, Winnipeg, MB, Canada
| | - Mahadevappa Hemshekhar
- Manitoba Centre for Proteomics and Systems Biology, Department of Internal Medicine, University of Manitoba, Winnipeg, MB, Canada
| | - Victor Spicer
- Department of Immunology, University of Toronto, Toronto, ON, Canada
| | - Anne M. van der Does
- PulmoScience Lab, Department of Pulmonology, Leiden University Medical Center, Leiden, The Netherlands
| | - Neeloffer Mookherjee
- Department of Immunology, University of Manitoba, Winnipeg, MB, Canada
- Manitoba Centre for Proteomics and Systems Biology, Department of Internal Medicine, University of Manitoba, Winnipeg, MB, Canada
| |
Collapse
|
172
|
So WY, Johnson B, Gordon PB, Bishop KS, Gong H, Burr HA, Staunton JR, Handler C, Sood R, Scarcelli G, Tanner K. Macrophage mediated mesoscale brain mechanical homeostasis mechanically imaged via optical tweezers and Brillouin microscopy in vivo. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2023.12.27.573380. [PMID: 38234798 PMCID: PMC10793422 DOI: 10.1101/2023.12.27.573380] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/19/2024]
Abstract
Tissues are active materials where epithelial turnover, immune surveillance, and remodeling of stromal cells such as macrophages all regulate form and function. Scattering modalities such as Brillouin microscopy (BM) can non-invasively access mechanical signatures at GHz. However, our traditional understanding of tissue material properties is derived mainly from modalities which probe mechanical properties at different frequencies. Thus, reconciling measurements amongst these modalities remains an active area. Here, we compare optical tweezer active microrheology (OT-AMR) and Brillouin microscopy (BM) to longitudinally map brain development in the larval zebrafish. We determine that each measurement is able to detect a mechanical signature linked to functional units of the brain. We demonstrate that the corrected BM-Longitudinal modulus using a density factor correlates well with OT-AMR storage modulus at lower frequencies. We also show that the brain tissue mechanical properties are dependent on both the neuronal architecture and the presence of macrophages. Moreover, the BM technique is able to delineate the contributions to mechanical properties of the macrophage from that due to colony stimulating factor 1 receptor (CSF1R) mediated stromal remodeling. Here, our data suggest that macrophage remodeling is instrumental in the maintenance of tissue mechanical homeostasis during development. Moreover, the strong agreement between the OT-AM and BM further demonstrates that scattering-based technique is sensitive to both large and minute structural modification in vivo.
Collapse
Affiliation(s)
- Woong Young So
- National Cancer Institute, National Institutes of Health (NIH), MD, USA
| | - Bailey Johnson
- National Cancer Institute, National Institutes of Health (NIH), MD, USA
| | | | - Kevin S. Bishop
- National Cancer Institute, National Institutes of Health (NIH), MD, USA
| | - Hyeyeon Gong
- National Cancer Institute, National Institutes of Health (NIH), MD, USA
- University of Maryland - College Park, MD, USA
| | - Hannah A Burr
- National Cancer Institute, National Institutes of Health (NIH), MD, USA
| | | | | | - Raman Sood
- National Human Genome Research Institute, NIH, MD, USA
| | | | - Kandice Tanner
- National Cancer Institute, National Institutes of Health (NIH), MD, USA
| |
Collapse
|
173
|
Lin X, Huang S, Gao S, Liu J, Tang J, Yu M. Integrin β5 subunit regulates hyperglycemia-induced vascular endothelial cell apoptosis through FoxO1-mediated macroautophagy. Chin Med J (Engl) 2024; 137:565-576. [PMID: 37500497 PMCID: PMC10932531 DOI: 10.1097/cm9.0000000000002769] [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: 01/25/2023] [Indexed: 07/29/2023] Open
Abstract
BACKGROUND Hyperglycemia frequently induces apoptosis in endothelial cells and ultimately contributes to microvascular dysfunction in patients with diabetes mellitus (DM). Previous research reported that the expression of integrins as well as their ligands was elevated in the diseased vessels of DM patients. However, the association between integrins and hyperglycemia-induced cell death is still unclear. This research was designed to investigate the role played by integrin subunit β5 (ITGB5) in hyperglycemia-induced endothelial cell apoptosis. METHODS We used leptin receptor knockout (Lepr-KO) ( db / db ) mice as spontaneous diabetes animal model. Selective deletion of ITGB5 in endothelial cell was achieved by injecting vascular targeted adeno-associated virus via tail vein. Besides, we also applied small interfering RNA in vitro to study the mechanism of ITGB5 in regulating high glucose-induced cell apoptosis. RESULTS ITGB5 and its ligand, fibronectin, were both upregulated after exposure to high glucose in vivo and in vitro . ITGB5 knockdown alleviated hyperglycemia-induced vascular endothelial cell apoptosis and microvascular rarefaction in vivo.In vitro analysis revealed that knockdown of either ITGB5 or fibronectin ameliorated high glucose-induced apoptosis in human umbilical vascular endothelial cells (HUVECs). In addition, knockdown of ITGB5 inhibited fibronectin-induced HUVEC apoptosis, which indicated that the fibronectin-ITGB5 interaction participated in high glucose-induced endothelial cell apoptosis. By using RNA-sequencing technology and bioinformatic analysis, we identified Forkhead Box Protein O1 (FoxO1) as an important downstream target regulated by ITGB5. Moreover, we demonstrated that the excessive macroautophagy induced by high glucose can contribute to HUVEC apoptosis, which was regulated by the ITGB5-FoxO1 axis. CONCLUSION The study revealed that high glucose-induced endothelial cell apoptosis was positively regulated by ITGB5, which suggested that ITGB5 could potentially be used to predict and treat DM-related vascular complications.
Collapse
Affiliation(s)
- Xuze Lin
- Department of Cardiology, Fuwai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Science and Peking Union Medical College, Beijing 100037, China
| | - Sizhuang Huang
- Department of Cardiology, Fuwai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Science and Peking Union Medical College, Beijing 100037, China
| | - Side Gao
- Department of Cardiology, Fuwai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Science and Peking Union Medical College, Beijing 100037, China
| | - Jinxing Liu
- Department of Cardiology, Fuwai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Science and Peking Union Medical College, Beijing 100037, China
| | - Jiong Tang
- Department of Cardiology, Fuwai Yunnan Cardiovascular Hospital, Kunming, Yunnan 650000, China
| | - Mengyue Yu
- Department of Cardiology, Fuwai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Science and Peking Union Medical College, Beijing 100037, China
| |
Collapse
|
174
|
Rizo G, Barrera AD, García EV, Roldán-Olarte M. Plasminogen activation and plasmin inhibition during in vitro fertilization in bovine: implications for fertilization parameters and early embryo development. Reprod Biol 2024; 24:100844. [PMID: 38160587 DOI: 10.1016/j.repbio.2023.100844] [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: 05/09/2023] [Revised: 11/24/2023] [Accepted: 12/14/2023] [Indexed: 01/03/2024]
Abstract
Components of the plasminogen/plasmin system, known to be present in the oocyte, play a key role in maturation and fertilization. The objective of this study was to examine the effect of plasminogen activation and plasmin inhibition by exogenous supplementation of the IVF medium with streptokinase (SK) or ɛ-aminocaproic acid (ε-ACA), respectively, on fertilization parameters and preimplantation embryo development. After in vitro maturation, bovine cumulus-oocyte complexes (COCs) were inseminated in the presence of SK or ε-ACA. The addition of SK to the IVF medium facilitated the adhesion of the spermatozoa to the zona pellucida without affecting the percentages of monospermy. Cleavage rates and blastocyst yield were similar between the SK and Control groups while they were lower with the ε-ACA treatment. Additionally, we found that the expression levels of embryo quality-related genes (SDHA and DNMT3A) could be modified in blastocysts by the addition of SK or ε-ACA during IVF. The results obtained indicate that supplementation of the IVF medium with SK did not greatly alter the embryonic developmental parameters related to embryo quality in blastocysts. Moreover, we noticed that ε-ACA treatment compromises the success of in vitro embryo development, thus highlighting the importance of the plasminogen/plasmin activity during the early stages of embryogenesis in bovine.
Collapse
Affiliation(s)
- Gabriela Rizo
- Instituto Superior de Investigaciones Biológicas (INSIBIO), CONICET-UNT, and Instituto de Biología 'Dr. Francisco D. Barbieri', Facultad de Bioquímica, Química y Farmacia, UNT, Chacabuco 461 (4000), San Miguel de Tucumán, Tucumán, Argentina
| | - Antonio Daniel Barrera
- Laboratorio de Biología Molecular, Facultad de Ciencias Agrarias y Veterinarias, Universidad Católica de Salta (UCASAL) and Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), Campo Castañares (4400), Salta, Argentina
| | - Elina Vanesa García
- Laboratorio de Biología Molecular, Facultad de Ciencias Agrarias y Veterinarias, Universidad Católica de Salta (UCASAL) and Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), Campo Castañares (4400), Salta, Argentina
| | - Mariela Roldán-Olarte
- Instituto Superior de Investigaciones Biológicas (INSIBIO), CONICET-UNT, and Instituto de Biología 'Dr. Francisco D. Barbieri', Facultad de Bioquímica, Química y Farmacia, UNT, Chacabuco 461 (4000), San Miguel de Tucumán, Tucumán, Argentina.
| |
Collapse
|
175
|
Egro FM, Schilling BK, Fisher JD, Saadoun R, Rubin JP, Marra KG, Solari MG. The Future of Microsurgery: Vascularized Composite Allotransplantation and Engineering Vascularized Tissue. J Hand Microsurg 2024; 16:100011. [PMID: 38854368 PMCID: PMC11127549 DOI: 10.1055/s-0042-1757182] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
Background Microsurgical techniques have revolutionized the field of reconstructive surgery and are the mainstay for complex soft tissue reconstruction. However, their limitations have promoted the development of viable alternatives. This article seeks to explore technologies that have the potential of revolutionizing microsurgical reconstruction as it is currently known, reflect on current and future vascularized composite allotransplantation (VCA) practices, as well as describe the basic science within emerging technologies and their potential translational applications. Methods A literature review was performed of the technologies that may represent the future of microsurgery: vascularized tissue engineering (VCA) and flap-specific tissue engineering. Results VCA has shown great promise and has already been employed in the clinical setting (especially in face and limb transplantation). Immunosuppression, logistics, cost, and regulatory pathways remain barriers to overcome to make it freely available. Vascularized and flap-specific tissue engineering remain a laboratory reality but have the potential to supersede VCA. The capability of creating an off-the-shelf free flap matching the required tissue, size, and shape is a significant advantage. However, these technologies are still at the early stage and require significant advancement before they can be translated into the clinical setting. Conclusion VCA, vascularized tissue engineering, and flap-specific bioengineering represent possible avenues for the evolution of current microsurgical techniques. The next decade will elucidate which of these three strategies will evolve into a tangible translational option and hopefully bring a paradigm shift of reconstructive surgery.
Collapse
Affiliation(s)
- Francesco M. Egro
- Department of Plastic Surgery, School of Medicine, University of Pittsburgh, Pittsburgh, Pennsylvania, United States
| | - Benjamin K. Schilling
- Department of Bioengineering, School of Engineering, University of Pittsburgh, Pittsburgh, Pennsylvania, United States
| | - James D. Fisher
- Department of Plastic Surgery, School of Medicine, University of Pittsburgh, Pittsburgh, Pennsylvania, United States
- Department of Bioengineering, School of Engineering, University of Pittsburgh, Pittsburgh, Pennsylvania, United States
| | - Rakan Saadoun
- Department of Plastic Surgery, School of Medicine, University of Pittsburgh, Pittsburgh, Pennsylvania, United States
| | - J. Peter Rubin
- Department of Plastic Surgery, School of Medicine, University of Pittsburgh, Pittsburgh, Pennsylvania, United States
- Department of Bioengineering, School of Engineering, University of Pittsburgh, Pittsburgh, Pennsylvania, United States
- McGowan Institute for Regenerative Medicine, University of Pittsburgh, Pittsburgh, Pennsylvania, United States
| | - Kacey G. Marra
- Department of Plastic Surgery, School of Medicine, University of Pittsburgh, Pittsburgh, Pennsylvania, United States
- Department of Bioengineering, School of Engineering, University of Pittsburgh, Pittsburgh, Pennsylvania, United States
- McGowan Institute for Regenerative Medicine, University of Pittsburgh, Pittsburgh, Pennsylvania, United States
| | - Mario G. Solari
- Department of Plastic Surgery, School of Medicine, University of Pittsburgh, Pittsburgh, Pennsylvania, United States
- McGowan Institute for Regenerative Medicine, University of Pittsburgh, Pittsburgh, Pennsylvania, United States
| |
Collapse
|
176
|
Singh M, Jassal R, Khetarpal P. Diagnostic and therapeutic approaches for endometriosis: a patent landscape. Arch Gynecol Obstet 2024; 309:831-842. [PMID: 37626175 DOI: 10.1007/s00404-023-07151-0] [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: 01/13/2023] [Accepted: 07/09/2023] [Indexed: 08/27/2023]
Abstract
OBJECTIVE The aim of this review is to analyze the patent filings and to systematize the main technological trends in patent protection for the diagnosis and therapeutics for endometriosis. Patent literature has also been explored to identify active inventors and applicants in this field. METHODOLOGY Patent search was carried out in the freely accessible patent search databases namely, patentscope using various combinations of the keywords "Endometriosis OR Adenomyosis" AND "Diagnostic OR Therapeutics" were used along with wildcard search queries in the "Title", "Abstract" and "Descriptions" fields. RESULTS A patent search revealed 144 patents describing inventions for diagnostic and therapeutic purposes of endometriosis. These patents include 26 patent applications in the diagnostic utility and 116 patent applications under the therapeutic approaches. Out of these 116 patent applications, 43 describe traditional medicines for endometriosis. Two patent applications describe inventions that can fall into both categories. CONCLUSION Efforts are being made to improve current diagnostic instruments. Hormonal alteration methods is the most common field of invention, followed by surgical interventions for therapeutics. A general trend of increase in patent application filings has been observed with a slight decrease in recent years.
Collapse
Affiliation(s)
- Maninder Singh
- Department of Human Genetics and Molecular Medicine, Central University of Punjab, Bathinda, 151401, India
| | - Reena Jassal
- Department of Pharmaceutical Sciences and Drug Research, Punjabi University, Patiala, 147002, India
| | - Preeti Khetarpal
- Human Genetics and Molecular Medicine Department, Central University of Punjab, Bathinda, 151001, India.
| |
Collapse
|
177
|
Rani A, Pandey DM, Pandey JP. Biomolecular characterization of Antheraea mylitta cocoonase: A secreted protease. Anal Biochem 2024; 686:115408. [PMID: 38008303 DOI: 10.1016/j.ab.2023.115408] [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: 09/10/2023] [Revised: 11/16/2023] [Accepted: 11/20/2023] [Indexed: 11/28/2023]
Abstract
Cocoonase is a protease secreted during the emergence of silk moths. In the present study cocoonase of Antheraea mylitta was collected, purified and secondary structure was determined using circular dichroism (CD) spectroscopy which revealed the presence of α-helix 4.3%, β-sheet 55%, turn 8% and random coil 32.7%. The thermal stability of cocoonase was studied using CD spectroscopy while the thermal property was observed using Differential Scanning Calorimetry (DSC). Furthermore, MALDI-TOF peptide mass fingerprinting (PMF) was performed for similar protein identification using the MASCOT server. Using casein as the substrate, the kinetic constants Km and Vmax were 13 × 103 mg/ml and 15.09 × 10-2 μg/mg.s1 respectively. The specific activity of cocoonase was observed to be maximum at temperature 40 °C, pH-8.0. The effect of heavy metals Hg2+, Cd2+, Co2+, Pb2+ showed inhibitory activity at higher concentrations, while few metals like Mn2+, Fe3+ enhanced the activity while the effect of Ca2+ was not much on the activity. Soybean trypsin inhibitor and PMSF showed an inhibitory effect on the activity of cocoonase. Additionally, antioxidant scavenging and fibrinolytic properties were also observed. Furthermore, the imperative information generated through the present study will serve to explore cocoonase for its prospective pharmaceutical applications.
Collapse
Affiliation(s)
- Aruna Rani
- Birla Institute of Technology, Mesra, Ranchi, 835215, Jharkhand, India; Central Tasar Research and Training Institute (Central Silk Board, MOT Govt. of India), Piska Nagri, Ranchi, 835303, Jharkhand, India
| | - Dev Mani Pandey
- Birla Institute of Technology, Mesra, Ranchi, 835215, Jharkhand, India
| | - Jay Prakash Pandey
- Central Tasar Research and Training Institute (Central Silk Board, MOT Govt. of India), Piska Nagri, Ranchi, 835303, Jharkhand, India.
| |
Collapse
|
178
|
Crossley RM, Johnson S, Tsingos E, Bell Z, Berardi M, Botticelli M, Braat QJS, Metzcar J, Ruscone M, Yin Y, Shuttleworth R. Modeling the extracellular matrix in cell migration and morphogenesis: a guide for the curious biologist. Front Cell Dev Biol 2024; 12:1354132. [PMID: 38495620 PMCID: PMC10940354 DOI: 10.3389/fcell.2024.1354132] [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: 12/11/2023] [Accepted: 02/12/2024] [Indexed: 03/19/2024] Open
Abstract
The extracellular matrix (ECM) is a highly complex structure through which biochemical and mechanical signals are transmitted. In processes of cell migration, the ECM also acts as a scaffold, providing structural support to cells as well as points of potential attachment. Although the ECM is a well-studied structure, its role in many biological processes remains difficult to investigate comprehensively due to its complexity and structural variation within an organism. In tandem with experiments, mathematical models are helpful in refining and testing hypotheses, generating predictions, and exploring conditions outside the scope of experiments. Such models can be combined and calibrated with in vivo and in vitro data to identify critical cell-ECM interactions that drive developmental and homeostatic processes, or the progression of diseases. In this review, we focus on mathematical and computational models of the ECM in processes such as cell migration including cancer metastasis, and in tissue structure and morphogenesis. By highlighting the predictive power of these models, we aim to help bridge the gap between experimental and computational approaches to studying the ECM and to provide guidance on selecting an appropriate model framework to complement corresponding experimental studies.
Collapse
Affiliation(s)
- Rebecca M. Crossley
- Wolfson Centre for Mathematical Biology, Mathematical Institute, University of Oxford, Oxford, United Kingdom
| | - Samuel Johnson
- Wolfson Centre for Mathematical Biology, Mathematical Institute, University of Oxford, Oxford, United Kingdom
| | - Erika Tsingos
- Computational Developmental Biology Group, Institute of Biodynamics and Biocomplexity, Utrecht University, Utrecht, Netherlands
| | - Zoe Bell
- Northern Institute for Cancer Research, Newcastle University, Newcastle upon Tyne, United Kingdom
| | - Massimiliano Berardi
- LaserLab, Department of Physics and Astronomy, Vrije Universiteit Amsterdam, Amsterdam, Netherlands
- Optics11 life, Amsterdam, Netherlands
| | | | - Quirine J. S. Braat
- Department of Applied Physics and Science Education, Eindhoven University of Technology, Eindhoven, Netherlands
| | - John Metzcar
- Department of Intelligent Systems Engineering, Indiana University, Bloomington, IN, United States
- Department of Informatics, Indiana University, Bloomington, IN, United States
| | | | - Yuan Yin
- Wolfson Centre for Mathematical Biology, Mathematical Institute, University of Oxford, Oxford, United Kingdom
| | | |
Collapse
|
179
|
Raps S, Bahr L, Karkossa I, Rossol M, von Bergen M, Schubert K. Triclosan and its alternatives, especially chlorhexidine, modulate macrophage immune response with distinct modes of action. THE SCIENCE OF THE TOTAL ENVIRONMENT 2024; 914:169650. [PMID: 38159774 DOI: 10.1016/j.scitotenv.2023.169650] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/03/2023] [Revised: 12/22/2023] [Accepted: 12/22/2023] [Indexed: 01/03/2024]
Abstract
Since European regulators restricted the use of bacteriocidic triclosan (TCS), alternatives for TCS are emerging. Recently, TCS has been shown to reprogram immune metabolism, trigger the NLRP3 inflammasome, and subsequently the release of IL-1β in human macrophages, but data on substitutes is scarce. Hence, we aimed to examine the effects of TCS compared to its alternatives at the molecular level in human macrophages. LPS-stimulated THP-1 macrophages were exposed to TCS or its substitutes, including benzalkonium chloride, benzethonium chloride, chloroxylenol, chlorhexidine (CHX) and cetylpyridinium chloride, with the inhibitory concentration (IC10-value) of cell viability to decipher their mode of action. TCS induced the release of the pro-inflammatory cytokine TNF and high level of IL-1β, suggesting the activation of the NLRP3-inflammasome, which was confirmed by non-apparent IL-1β under the NLRP3-inhibitor MCC950 treatment d. While IL-6 release was reduced in all treatments, the alternative CHX completely abolished the release of all investigated cytokines. To unravel the underlying molecular mechanisms, we used untargeted LC-MS/MS-based proteomics. TCS and CHX showed the strongest cellular response at the protein and signalling pathway level, whereby pathways related to metabolism, translation, cellular stress and migration were mainly affected but to different proposed modes of action. TCS inhibited mitochondrial electron transfer and affected phagocytosis. In contrast, in CHX-treated cells, the translation was arrested due to stress conditions, resulting in the formation of stress granules. Mitochondrial (e.g. ATP5F1D, ATP5PB, UQCRQ) and ribosomal (e.g. RPL10, RPL35, RPS23) proteins were revealed as putative key drivers. Furthermore, we have demonstrated the formation of podosomes by CHX, potentially involved in ECM degradation. Our results exhibit modulation of the immune response in macrophages by TCS and its substitutes and illuminated underlying molecular effects. These results illustrate critical processes involved in the modulation of macrophages' immune response by TCS and its alternatives, providing information essential for hazard assessment.
Collapse
Affiliation(s)
- Stefanie Raps
- Department of Molecular Toxicology, Helmholtz-Centre for Environmental Research - UFZ, Leipzig, Germany
| | - Laura Bahr
- Department of Molecular Toxicology, Helmholtz-Centre for Environmental Research - UFZ, Leipzig, Germany
| | - Isabel Karkossa
- Department of Molecular Toxicology, Helmholtz-Centre for Environmental Research - UFZ, Leipzig, Germany
| | - Manuela Rossol
- Molecular Immunology, Faculty of Health Sciences, Brandenburg University of Technology Cottbus-Senftenberg, Germany
| | - Martin von Bergen
- Department of Molecular Toxicology, Helmholtz-Centre for Environmental Research - UFZ, Leipzig, Germany; Institute of Biochemistry, Leipzig University, Leipzig, Germany; German Centre for Integrative Biodiversity Research (iDiv) Halle-Jena-Leipzig, Leipzig, Germany
| | - Kristin Schubert
- Department of Molecular Toxicology, Helmholtz-Centre for Environmental Research - UFZ, Leipzig, Germany.
| |
Collapse
|
180
|
Ahmad M, Haffner-Luntzer M, Schoppa A, Najafova Z, Lukic T, Yorgan TA, Amling M, Schinke T, Ignatius A. Mechanical induction of osteoanabolic Wnt1 promotes osteoblast differentiation via Plat. FASEB J 2024; 38:e23489. [PMID: 38407813 DOI: 10.1096/fj.202301424rr] [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: 07/13/2023] [Revised: 01/04/2024] [Accepted: 01/30/2024] [Indexed: 02/27/2024]
Abstract
Physical activity-induced mechanical stimuli play a crucial role in preserving bone mass and structure by promoting bone formation. While the Wnt pathway is pivotal for mediating the osteoblast response to loading, the exact mechanisms are not fully understood. Here, we found that mechanical stimulation induces osteoblastic Wnt1 expression, resulting in an upregulation of key osteogenic marker genes, including Runx2 and Sp7, while Wnt1 knockdown using siRNA prevented these effects. RNAseq analysis identified Plat as a major target through which Wnt1 exerts its osteogenic influence. This was corroborated by Plat depletion using siRNA, confirming its positive role in osteogenic differentiation. Moreover, we demonstrated that mechanical stimulation enhances Plat expression, which, in turn leads to increased expression of osteogenic markers like Runx2 and Sp7. Notably, Plat depletion by siRNA prevented this effect. We have established that Wnt1 regulates Plat expression by activating β-Catenin. Silencing Wnt1 impairs mechanically induced β-Catenin activation, subsequently reducing Plat expression. Furthermore, our findings showed that Wnt1 is essential for osteoblasts to respond to mechanical stimulation and induce Runx2 and Sp7 expression, in part through the Wnt1/β-Catenin/Plat signaling pathway. Additionally, we observed significantly reduced Wnt1 and Plat expression in bones from ovariectomy (OVX)-induced and age-related osteoporotic mouse models compared with non-OVX and young mice, respectively. Overall, our data suggested that Wnt1 and Plat play significant roles in mechanically induced osteogenesis. Their decreased expression in bones from OVX and aged mice highlights their potential involvement in post-menopausal and age-related osteoporosis, respectively.
Collapse
Affiliation(s)
- Mubashir Ahmad
- Institute of Orthopedic Research and Biomechanics, University Medical Center Ulm, Ulm, Germany
| | - Melanie Haffner-Luntzer
- Institute of Orthopedic Research and Biomechanics, University Medical Center Ulm, Ulm, Germany
| | - Astrid Schoppa
- Institute of Orthopedic Research and Biomechanics, University Medical Center Ulm, Ulm, Germany
| | | | - Teodora Lukic
- Robert Bosch Center for Tumor Diseases, Stuttgart, Germany
| | - Timur Alexander Yorgan
- Department of Osteology and Biomechanics, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Michael Amling
- Department of Osteology and Biomechanics, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Thorsten Schinke
- Department of Osteology and Biomechanics, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Anita Ignatius
- Institute of Orthopedic Research and Biomechanics, University Medical Center Ulm, Ulm, Germany
| |
Collapse
|
181
|
Ganguly A, Mukherjee S, Chatterjee K, Spada S. Factors affecting heterogeneity in breast cancer microenvironment: A narrative mini review. INTERNATIONAL REVIEW OF CELL AND MOLECULAR BIOLOGY 2024; 385:211-226. [PMID: 38663960 DOI: 10.1016/bs.ircmb.2024.01.002] [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: 06/15/2024]
Abstract
Breast cancer (BC) heterogeneity is a key trait of BC tumors with crucial implications on tumorigenesis, diagnosis, and therapeutic modalities. It is influenced by tumor intrinsic features and by the tumor microenvironment (TME) composition of different intra-tumoral regions, which in turn affect cancer progression within patients. In this mini review, we will highlight the mechanisms that generate cancer heterogeneity in BC and how they affect the responses to cancer therapies.
Collapse
Affiliation(s)
- Anirban Ganguly
- Department of Biochemistry, All India Institute of Medical Sciences, Deoghar, India
| | - Sumit Mukherjee
- Department of Cardiothoracic and Vascular Surgery, Albert Einstein College of Medicine, Bronx, NY, United States
| | | | - Sheila Spada
- Tumor Immunology and Immunotherapy Unit, IRCCS Regina Elena National Cancer Institute, Rome, Italy.
| |
Collapse
|
182
|
Da W, Jiang W, Tao L. ROS/MMP-9 mediated CS degradation in BMSC inhibits citric acid metabolism participating in the dual regulation of bone remodelling. Cell Death Discov 2024; 10:77. [PMID: 38355572 PMCID: PMC10866869 DOI: 10.1038/s41420-024-01835-5] [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: 06/05/2023] [Revised: 01/27/2024] [Accepted: 01/29/2024] [Indexed: 02/16/2024] Open
Abstract
It is necessary to figure out the abnormal energy metabolites at the cellular level of postmenopausal osteoporosis (PMOP) bone microenvironment. In this study, we constructed PMOP model by ovariectomy and identified 9 differential metabolites compared with control femur by energy metabolomic. The enrichment analysis of differential metabolites revealed that tricarboxylic acid cycle, glucagon pathway and purinergic signaling pathway were the main abnormal metabolic processes. Citric acid was identified as the key metabolite by constructing compound reaction-enzyme-gene network. The functional annotation of citric acid targets identified by network pharmacological tools indicated that matrix metalloproteinase 9 (MMP-9) may be involved in regulating citric acid metabolism in the osteogenic differentiation of bone marrow mesenchymal stem cell (BMSC). Molecular docking shows that the interaction forces between MMP-9 and citric acid synthase (CS) is -638, and there are multiple groups of residues used to form hydrogen bonds. Exogenous H2O2 promotes the expression of MMP-9 in BMSC to further degrade CS resulting in a decrease in mitochondrial citric acid synthesis, which leads to the disorder of bone remodeling by two underlying mechanisms ((1) the decreased histone acetylation inhibits the osteogenic differentiation potential of BMSC; (2) the decreased bone mineralization by citric acid deposition). MMP-9-specific inhibitor (MMP-9-IN-1) could significantly improve the amount of CS in BMSC to promote cellular citric acid synthesis, and further enhance bone remodeling. These findings suggest inhibiting the degradation of CS by MMP-9 to promote the net production of citric acid in osteogenic differentiation of BMSC may be a new direction of PMOP research.
Collapse
Affiliation(s)
- Wacili Da
- Department of Orthopedics Surgery, Orthopedic Research Institute, West China Hospital, West China Medical School, Sichuan University, Chengdu, Sichuan Province, China
| | - Wen Jiang
- Department of Orthopedics, First Hospital of China Medical University, Shenyang, Liaoning, China.
| | - Lin Tao
- Department of Orthopedics, First Hospital of China Medical University, Shenyang, Liaoning, China.
| |
Collapse
|
183
|
Li Z, Yang X, Li Z. Tumor Mechanics Meets Nanomedicine Mechanical Properties. CHEMISTRY OF MATERIALS 2024; 36:1041-1053. [DOI: 10.1021/acs.chemmater.3c02474] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 03/28/2025]
Affiliation(s)
- Zheng Li
- Department of Nanomedicine and Biopharmaceuticals, College of Life Science and Technology, Huazhong University of Science and Technology, Wuhan, 430074, People’s Republic of China
| | - Xiangliang Yang
- Department of Nanomedicine and Biopharmaceuticals, College of Life Science and Technology, Huazhong University of Science and Technology, Wuhan, 430074, People’s Republic of China
- National Engineering Research Center for Nanomedicine, Huazhong University of Science and Technology, Wuhan, 430074, People’s Republic of China
- Key Laboratory of Molecular Biophysics of Ministry of Education, Huazhong University of Science and Technology, Wuhan, 430074, People’s Republic of China
- Hubei Key Laboratory of Bioinorganic Chemistry and Materia Medical, Huazhong University of Science and Technology, Wuhan, 430074, People’s Republic of China
- Hubei Engineering Research Center for Biomaterials and Medical Protective Materials, Huazhong University of Science and Technology, Wuhan, 430074, People’s Republic of China
| | - Zifu Li
- Department of Nanomedicine and Biopharmaceuticals, College of Life Science and Technology, Huazhong University of Science and Technology, Wuhan, 430074, People’s Republic of China
- National Engineering Research Center for Nanomedicine, Huazhong University of Science and Technology, Wuhan, 430074, People’s Republic of China
- Key Laboratory of Molecular Biophysics of Ministry of Education, Huazhong University of Science and Technology, Wuhan, 430074, People’s Republic of China
- Hubei Key Laboratory of Bioinorganic Chemistry and Materia Medical, Huazhong University of Science and Technology, Wuhan, 430074, People’s Republic of China
- Hubei Engineering Research Center for Biomaterials and Medical Protective Materials, Huazhong University of Science and Technology, Wuhan, 430074, People’s Republic of China
| |
Collapse
|
184
|
Abdallah R, Shaito AA, Badran A, Baydoun S, Sobeh M, Ouchari W, Sahri N, Eid AH, Mesmar JE, Baydoun E. Fractionation and phytochemical composition of an ethanolic extract of Ziziphus nummularia leaves: antioxidant and anticancerous properties in human triple negative breast cancer cells. Front Pharmacol 2024; 15:1331843. [PMID: 38405665 PMCID: PMC10885810 DOI: 10.3389/fphar.2024.1331843] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2023] [Accepted: 01/09/2024] [Indexed: 02/27/2024] Open
Abstract
Natural products have long been utilized in traditional medicine as remedies to improve health and treat illnesses, and have had a key role in modern drug discovery. Recently, there has been a revived interest in the search for bioactives from natural sources as alternative or complementary modalities to synthetic medicines; especially for cancer treatment, which incidence and mortality rates are on the rise worldwide. Ziziphus nummularia has been widely used in traditional medicine for the treatment of various diseases. Its traditional uses and numerous ethnopharmacological properties may be attributed to its richness in bioactive metabolites. However, its phytochemical composition or chemopreventive effects against the aggressive triple-negative breast cancer (TNBC) are still poorly explored. Here, phytochemical composition of an ethanolic extract of Z. nummularia leaves (ZNE) and its chromatographically isolated fractions was identified both qualitatively by spectrophotometric assays and analytically by HPLC-PDA-MS/MS. The anti-proliferative effects of ZNE were tested in several cancer cell lines, but we focused on its anti-TNBC effects since they were not explored yet. The anti-cancerous potential of ZNE and its fractions was tested in vitro in MDA-MB-231, a TNBC cell line. Results showed that ZNE and its Fraction 6 (F6) reduced the viability of MDA-MB-231 cells. F6 decreased MDA-MB-231 viability more than crude ZNE or its other fractions. ZNE and F6 are rich in phytochemicals and HPLC-PDA-MS/MS analysis identified several metabolites that were previously reported to have anti-cancerous effects. Both ZNE and F6 showed potent antioxidant capacity in the DPPH assay, but promoted reactive oxygen species (ROS) production in MDA-MB-231 cells; an effect which was blunted by the antioxidant N-acetyl cysteine (NAC). NAC also blunted ZNE- and F6-induced reduction in TNBC cell viability. We also demonstrated that ZNE and F6 induced an arrest of the cell cycle, and triggered apoptosis- and autophagy-mediated cell death. ZNE and F6 inhibited metastasis-related cellular processes by modifying cell migration, invasion, and adhesion. Taken together, our findings reveal that Z. nummularia is rich in phytochemicals that can attenuate the malignant phenotype of TNBC and may offer innovative avenues for the discovery of new drug leads for treatment of TNBC and other cancers.
Collapse
Affiliation(s)
- Rola Abdallah
- Department of Biology, American University of Beirut, Beirut, Lebanon
| | - Abdullah A. Shaito
- Biomedical Research Center, Department of Biomedical Sciences at College of Health Sciences, and College of Medicine, Qatar University, Doha, Qatar
| | - Adnan Badran
- Department of Nutrition, University of Petra, Amman, Jordan
| | - Serine Baydoun
- Breast Imaging Section, Imaging Institute, Cleveland Clinic Foundation, Cleveland, OH, United States
| | - Mansour Sobeh
- Department of Biology, American University of Beirut, Beirut, Lebanon
| | - Wafae Ouchari
- Department of Biology, American University of Beirut, Beirut, Lebanon
| | - Nihad Sahri
- Agrobiosciences Program, College for Agriculture and Environmental Science, Mohammed VI Polytechnic University, Ben Guerir, Morocco
| | - Ali H. Eid
- Department of Basic Medical Sciences, College of Medicine, QU Health, Qatar University, Doha, Qatar
| | | | - Elias Baydoun
- Department of Biology, American University of Beirut, Beirut, Lebanon
| |
Collapse
|
185
|
Kolahi Azar H, Gharibshahian M, Rostami M, Mansouri V, Sabouri L, Beheshtizadeh N, Rezaei N. The progressive trend of modeling and drug screening systems of breast cancer bone metastasis. J Biol Eng 2024; 18:14. [PMID: 38317174 PMCID: PMC10845631 DOI: 10.1186/s13036-024-00408-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2023] [Accepted: 01/22/2024] [Indexed: 02/07/2024] Open
Abstract
Bone metastasis is considered as a considerable challenge for breast cancer patients. Various in vitro and in vivo models have been developed to examine this occurrence. In vitro models are employed to simulate the intricate tumor microenvironment, investigate the interplay between cells and their adjacent microenvironment, and evaluate the effectiveness of therapeutic interventions for tumors. The endeavor to replicate the latency period of bone metastasis in animal models has presented a challenge, primarily due to the necessity of primary tumor removal and the presence of multiple potential metastatic sites.The utilization of novel bone metastasis models, including three-dimensional (3D) models, has been proposed as a promising approach to overcome the constraints associated with conventional 2D and animal models. However, existing 3D models are limited by various factors, such as irregular cellular proliferation, autofluorescence, and changes in genetic and epigenetic expression. The imperative for the advancement of future applications of 3D models lies in their standardization and automation. The utilization of artificial intelligence exhibits the capability to predict cellular behavior through the examination of substrate materials' chemical composition, geometry, and mechanical performance. The implementation of these algorithms possesses the capability to predict the progression and proliferation of cancer. This paper reviewed the mechanisms of bone metastasis following primary breast cancer. Current models of breast cancer bone metastasis, along with their challenges, as well as the future perspectives of using these models for translational drug development, were discussed.
Collapse
Affiliation(s)
- Hanieh Kolahi Azar
- Department of Pathology, Tabriz University of Medical Sciences, Tabriz, Iran
- Regenerative Medicine Group (REMED), Universal Scientific Education and Research Network (USERN), Tehran, Iran
| | - Maliheh Gharibshahian
- Department of Tissue Engineering, School of Medicine, Shahroud University of Medical Sciences, Shahroud, Iran
- Regenerative Medicine Group (REMED), Universal Scientific Education and Research Network (USERN), Tehran, Iran
| | - Mohammadreza Rostami
- Division of Food Safety and Hygiene, Department of Environmental Health Engineering, School of Public Health, Tehran University of Medical Sciences, Tehran, Iran
- Food Science and Nutrition Group (FSAN), Universal Scientific Education and Research Network (USERN), Tehran, Iran
| | - Vahid Mansouri
- Gene Therapy Research Center, Digestive Diseases Research Institute, Shariati Hospital, Tehran University of Medical Sciences, Tehran, Iran
- Regenerative Medicine Group (REMED), Universal Scientific Education and Research Network (USERN), Tehran, Iran
| | - Leila Sabouri
- Department of Tissue Engineering and Applied Cell Sciences, School of Paramedicine, Guilan University of Medical Sciences, Rasht, Iran
- Regenerative Medicine Group (REMED), Universal Scientific Education and Research Network (USERN), Tehran, Iran
| | - Nima Beheshtizadeh
- Department of Tissue Engineering, Faculty of Advanced Medical Sciences, Tabriz University of Medical Sciences, Tabriz, Iran.
- Regenerative Medicine Group (REMED), Universal Scientific Education and Research Network (USERN), Tehran, Iran.
| | - Nima Rezaei
- Department of Immunology, School of Medicine, Tehran University of Medical Sciences, Tehran, Iran.
- Research Center for Immunodeficiencies, Children's Medical Center, Tehran University of Medical Sciences, Tehran, Iran.
- Network of Immunity in Infection, Malignancy and Autoimmunity (NIIMA), Universal Scientific Education and Research Network (USERN), Tehran, Iran.
| |
Collapse
|
186
|
Fan X, Sun AR, Young RSE, Afara IO, Hamilton BR, Ong LJY, Crawford R, Prasadam I. Spatial analysis of the osteoarthritis microenvironment: techniques, insights, and applications. Bone Res 2024; 12:7. [PMID: 38311627 PMCID: PMC10838951 DOI: 10.1038/s41413-023-00304-6] [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: 09/05/2023] [Revised: 11/21/2023] [Accepted: 11/27/2023] [Indexed: 02/06/2024] Open
Abstract
Osteoarthritis (OA) is a debilitating degenerative disease affecting multiple joint tissues, including cartilage, bone, synovium, and adipose tissues. OA presents diverse clinical phenotypes and distinct molecular endotypes, including inflammatory, metabolic, mechanical, genetic, and synovial variants. Consequently, innovative technologies are needed to support the development of effective diagnostic and precision therapeutic approaches. Traditional analysis of bulk OA tissue extracts has limitations due to technical constraints, causing challenges in the differentiation between various physiological and pathological phenotypes in joint tissues. This issue has led to standardization difficulties and hindered the success of clinical trials. Gaining insights into the spatial variations of the cellular and molecular structures in OA tissues, encompassing DNA, RNA, metabolites, and proteins, as well as their chemical properties, elemental composition, and mechanical attributes, can contribute to a more comprehensive understanding of the disease subtypes. Spatially resolved biology enables biologists to investigate cells within the context of their tissue microenvironment, providing a more holistic view of cellular function. Recent advances in innovative spatial biology techniques now allow intact tissue sections to be examined using various -omics lenses, such as genomics, transcriptomics, proteomics, and metabolomics, with spatial data. This fusion of approaches provides researchers with critical insights into the molecular composition and functions of the cells and tissues at precise spatial coordinates. Furthermore, advanced imaging techniques, including high-resolution microscopy, hyperspectral imaging, and mass spectrometry imaging, enable the visualization and analysis of the spatial distribution of biomolecules, cells, and tissues. Linking these molecular imaging outputs to conventional tissue histology can facilitate a more comprehensive characterization of disease phenotypes. This review summarizes the recent advancements in the molecular imaging modalities and methodologies for in-depth spatial analysis. It explores their applications, challenges, and potential opportunities in the field of OA. Additionally, this review provides a perspective on the potential research directions for these contemporary approaches that can meet the requirements of clinical diagnoses and the establishment of therapeutic targets for OA.
Collapse
Affiliation(s)
- Xiwei Fan
- Centre for Biomedical Technologies, Queensland University of Technology, Brisbane, QLD, Australia
- School of Mechanical, Medical & Process Engineering, Queensland University of Technology, Brisbane, QLD, Australia
| | - Antonia Rujia Sun
- Centre for Biomedical Technologies, Queensland University of Technology, Brisbane, QLD, Australia
- School of Mechanical, Medical & Process Engineering, Queensland University of Technology, Brisbane, QLD, Australia
| | - Reuben S E Young
- Central Analytical Research Facility, Queensland University of Technology, Brisbane, QLD, Australia
- Molecular Horizons, University of Wollongong, Wollongong, NSW, Australia
| | - Isaac O Afara
- Department of Technical Physics, University of Eastern Finland, Kuopio, Finland
- School of Electrical Engineering and Computer Science, Faculty of Engineering, Architecture and Information Technology, University of Queensland, Brisbane, QLD, Australia
| | - Brett R Hamilton
- Centre for Microscopy and Microanalysis, University of Queensland, Brisbane, QLD, Australia
| | - Louis Jun Ye Ong
- Centre for Biomedical Technologies, Queensland University of Technology, Brisbane, QLD, Australia
- School of Mechanical, Medical & Process Engineering, Queensland University of Technology, Brisbane, QLD, Australia
| | - Ross Crawford
- Centre for Biomedical Technologies, Queensland University of Technology, Brisbane, QLD, Australia
- The Prince Charles Hospital, Brisbane, QLD, Australia
| | - Indira Prasadam
- Centre for Biomedical Technologies, Queensland University of Technology, Brisbane, QLD, Australia.
- School of Mechanical, Medical & Process Engineering, Queensland University of Technology, Brisbane, QLD, Australia.
| |
Collapse
|
187
|
Crawford AJ, Gomez-Cruz C, Russo GC, Huang W, Bhorkar I, Roy T, Muñoz-Barrutia A, Wirtz D, Garcia-Gonzalez D. Tumor proliferation and invasion are intrinsically coupled and unraveled through tunable spheroid and physics-based models. Acta Biomater 2024; 175:170-185. [PMID: 38160858 DOI: 10.1016/j.actbio.2023.12.043] [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: 09/28/2023] [Revised: 12/13/2023] [Accepted: 12/26/2023] [Indexed: 01/03/2024]
Abstract
Proliferation and invasion are two key drivers of tumor growth that are traditionally considered independent multicellular processes. However, these processes are intrinsically coupled through a maximum carrying capacity, i.e., the maximum spatial cell concentration supported by the tumor volume, total cell count, nutrient access, and mechanical properties of the tissue stroma. We explored this coupling of proliferation and invasion through in vitro and in silico methods where we modulated the mechanical properties of the tumor and the surrounding extracellular matrix. E-cadherin expression and stromal collagen concentration were manipulated in a tunable breast cancer spheroid to determine the overall impacts of these tumor variables on net tumor proliferation and continuum invasion. We integrated these results into a mixed-constitutive formulation to computationally delineate the influences of cellular and extracellular adhesion, stiffness, and mechanical properties of the extracellular matrix on net proliferation and continuum invasion. This framework integrates biological in vitro data into concise computational models of invasion and proliferation to provide more detailed physical insights into the coupling of these key tumor processes and tumor growth. STATEMENT OF SIGNIFICANCE: Tumor growth involves expansion into the collagen-rich stroma through intrinsic coupling of proliferation and invasion within the tumor continuum. These processes are regulated by a maximum carrying capacity that is determined by the total cell count, tumor volume, nutrient access, and mechanical properties of the surrounding stroma. The influences of biomechanical parameters (i.e., stiffness, cell elongation, net proliferation rate and cell-ECM friction) on tumor proliferation or invasion cannot be unraveled using experimental methods alone. By pairing a tunable spheroid system with computational modeling, we delineated the interdependencies of each system parameter on tumor proliferation and continuum invasion, and established a concise computational framework for studying tumor mechanobiology.
Collapse
Affiliation(s)
- Ashleigh J Crawford
- Department of Chemical and Biomolecular Engineering, Johns Hopkins University, 3400N Charles St, Baltimore, MD 21218, USA; Johns Hopkins Physical Sciences-Oncology Center and Institute for NanoBioTechnology, Johns Hopkins University, 3400N Charles St, Baltimore, Maryland 21218, USA
| | - Clara Gomez-Cruz
- Department of Continuum Mechanics and Structural Analysis, Universidad Carlos III de Madrid, Avda. de la Universidad 30, 28911 Leganes, Madrid, Spain; Departamento de Bioingenieria, Universidad Carlos III de Madrid, Avda. de la Universidad 30, 28911 Leganes, Madrid, Spain
| | - Gabriella C Russo
- Department of Chemical and Biomolecular Engineering, Johns Hopkins University, 3400N Charles St, Baltimore, MD 21218, USA; Johns Hopkins Physical Sciences-Oncology Center and Institute for NanoBioTechnology, Johns Hopkins University, 3400N Charles St, Baltimore, Maryland 21218, USA
| | - Wilson Huang
- Johns Hopkins Physical Sciences-Oncology Center and Institute for NanoBioTechnology, Johns Hopkins University, 3400N Charles St, Baltimore, Maryland 21218, USA; Department of Biology, Johns Hopkins University, 3400N Charles St, Baltimore, Maryland 21218, USA
| | - Isha Bhorkar
- Johns Hopkins Physical Sciences-Oncology Center and Institute for NanoBioTechnology, Johns Hopkins University, 3400N Charles St, Baltimore, Maryland 21218, USA; Department of Biomedical Engineering, Johns Hopkins University, 3400N Charles St, Baltimore, Maryland 21218, USA
| | - Triya Roy
- Department of Chemical and Biomolecular Engineering, Johns Hopkins University, 3400N Charles St, Baltimore, MD 21218, USA; Johns Hopkins Physical Sciences-Oncology Center and Institute for NanoBioTechnology, Johns Hopkins University, 3400N Charles St, Baltimore, Maryland 21218, USA
| | - Arrate Muñoz-Barrutia
- Department of Chemical and Biomolecular Engineering, Johns Hopkins University, 3400N Charles St, Baltimore, MD 21218, USA; Departamento de Bioingenieria, Universidad Carlos III de Madrid, Avda. de la Universidad 30, 28911 Leganes, Madrid, Spain; Area de Ingenieria Biomedica, Instituto de Investigacion Sanitaria Gregorio Maranon, Calle del Doctor Esquerdo 46, Madrid' ES 28007, Spain
| | - Denis Wirtz
- Department of Chemical and Biomolecular Engineering, Johns Hopkins University, 3400N Charles St, Baltimore, MD 21218, USA; Johns Hopkins Physical Sciences-Oncology Center and Institute for NanoBioTechnology, Johns Hopkins University, 3400N Charles St, Baltimore, Maryland 21218, USA; Department of Biomedical Engineering, Johns Hopkins University, 3400N Charles St, Baltimore, Maryland 21218, USA; Departments of Pathology and Oncology and Sydney Kimmel Comprehensive Cancer Center, The Johns Hopkins School of Medicine, 1800 Orleans St, Baltimore, MD 21215, USA.
| | - Daniel Garcia-Gonzalez
- Department of Continuum Mechanics and Structural Analysis, Universidad Carlos III de Madrid, Avda. de la Universidad 30, 28911 Leganes, Madrid, Spain.
| |
Collapse
|
188
|
Yamaura K, Nishimura H, Ruzbarsky JJ, Kuhns BD, Mullen MT, Duke VR, O’Hara KM, Brown JM, Comfort SM, Hambright WS, Bahney CS, Huard J, Philippon MJ. Evaluating the Morphology of Unique Superficial Fissured Cartilage Lesions at the Femoral Head-Neck Junction in Patients with Femoroacetabular Impingement Syndrome. Orthop J Sports Med 2024; 12:23259671231219217. [PMID: 38343646 PMCID: PMC10854385 DOI: 10.1177/23259671231219217] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/19/2023] [Accepted: 06/29/2023] [Indexed: 03/18/2025] Open
Abstract
Background While an association between femoroacetabular impingement (FAI) and osteoarthritis (OA) has been reported, the mechanistic differences and transition between the 2 conditions is not fully understood. In FAI, cartilage lesions at the femoral head-neck junction can sometimes be visualized during hip arthroscopy. Purpose/Hypothesis The purpose of this study was to describe a unique dimpled pattern of superficial fissured cartilage lesions on the femoral head-neck junction at impingement site in patients with FAI syndrome (FAIS) and to evaluate the clinical, histological, and genetic phenotype of this cartilage. We hypothesized that the cartilage lesions may indicate risk for, or predict occurrence of, OA. Study Design Controlled laboratory study. Methods Six hips (6 patients; mean age, 34.2 ± 12.9 years; range, 19-54 years) with dimpled or fissured cartilage were included among patients who underwent hip arthroscopy for treatment of FAIS from October 2020 through December 2021. This affected cartilage (dimple-pattern group) and normal cartilage (control group) on the femoral head-neck junction were collected from the same patients and evaluated for histological quantification by Mankin scores and expression of proteins related to cartilage degeneration (eg, matrix metalloproteinase [MMP]-1, MMP-2, MMP-3, MMP-10, and MMP-12, tissue inhibitor of metalloproteinase [TIMP]-1 and TMP-2, aggrecan neopepitope CS846, and hyaluronic acid [HA]) with the use of Milliplex Multiplex Assays. Results All 6 hips were of the mixed FAI subtype. Preoperatively, 4 of 6 hips had Tönnis grade 1 radiographic changes, which was associated with greater femoral head chondral damage visualized intraoperatively. Mankin scores for the normal cartilage group and the dimple-pattern group were 0.67 ± 0.82 and 3.3 ± 0.82, respectively. Dimple pattern fissured cartilage showed a significant increase in Mankin score (P = .031) and a significant increase in protein expression of CS846 (P = .031) compared with normal cartilage. There were no significant differences in MMPs, TIMPs, or HA levels between the 2 groups. Conclusion The dimple pattern fissured cartilage, compared to normal cartilage, showed histologically significant cartilage degeneration and a significant increase in protein expression of CS846, a biomarker for early OA. Clinical Relevance This lesion serves as helpful visual indicator of early degeneration of the cartilage of femoral head-neck junction caused by FAIS.
Collapse
Affiliation(s)
- Kohei Yamaura
- Center for Regenerative and Personalized Medicine, Steadman Philippon Research Institute, Vail, Colorado, USA
- Department of Orthopaedic Surgery, Kobe University Graduate School of Medicine, Kobe, Japan
| | - Haruki Nishimura
- Center for Regenerative and Personalized Medicine, Steadman Philippon Research Institute, Vail, Colorado, USA
| | - Joseph J. Ruzbarsky
- Center for Regenerative and Personalized Medicine, Steadman Philippon Research Institute, Vail, Colorado, USA
- The Steadman Clinic, Vail, Colorado, USA
| | - Benjamin D. Kuhns
- Center for Regenerative and Personalized Medicine, Steadman Philippon Research Institute, Vail, Colorado, USA
- The Steadman Clinic, Vail, Colorado, USA
| | - Michael T. Mullen
- Center for Regenerative and Personalized Medicine, Steadman Philippon Research Institute, Vail, Colorado, USA
| | - Victoria R. Duke
- Center for Regenerative and Personalized Medicine, Steadman Philippon Research Institute, Vail, Colorado, USA
| | - Kelsey M. O’Hara
- Center for Regenerative and Personalized Medicine, Steadman Philippon Research Institute, Vail, Colorado, USA
| | - Jarrod M. Brown
- Center for Regenerative and Personalized Medicine, Steadman Philippon Research Institute, Vail, Colorado, USA
| | - Spencer M. Comfort
- Center for Regenerative and Personalized Medicine, Steadman Philippon Research Institute, Vail, Colorado, USA
| | - William S. Hambright
- Center for Regenerative and Personalized Medicine, Steadman Philippon Research Institute, Vail, Colorado, USA
| | - Chelsea S. Bahney
- Center for Regenerative and Personalized Medicine, Steadman Philippon Research Institute, Vail, Colorado, USA
- The Orthopaedic Trauma Institute, University of California, San Francisco (UCSF), San Francisco, California, USA
| | - Johnny Huard
- Center for Regenerative and Personalized Medicine, Steadman Philippon Research Institute, Vail, Colorado, USA
| | - Marc J. Philippon
- Center for Regenerative and Personalized Medicine, Steadman Philippon Research Institute, Vail, Colorado, USA
- The Steadman Clinic, Vail, Colorado, USA
| |
Collapse
|
189
|
Caputo A, Vipparthi K, Bazeley P, Downs-Kelly E, McIntire P, Duckworth LA, Ni Y, Hu B, Keri RA, Karaayvaz M. Spatial Transcriptomics Suggests That Alterations Occur in the Preneoplastic Breast Microenvironment of BRCA1/2 Mutation Carriers. Mol Cancer Res 2024; 22:169-180. [PMID: 37878345 PMCID: PMC10872731 DOI: 10.1158/1541-7786.mcr-23-0489] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2023] [Revised: 08/28/2023] [Accepted: 10/23/2023] [Indexed: 10/26/2023]
Abstract
Breast cancer is the most common cancer in females, affecting one in every eight women and accounting for the majority of cancer-related deaths in women worldwide. Germline mutations in the BRCA1 and BRCA2 genes are significant risk factors for specific subtypes of breast cancer. BRCA1 mutations are associated with basal-like breast cancers, whereas BRCA2 mutations are associated with luminal-like disease. Defects in mammary epithelial cell differentiation have been previously recognized in germline BRCA1/2 mutation carriers even before cancer incidence. However, the underlying mechanism is largely unknown. Here, we employ spatial transcriptomics to investigate defects in mammary epithelial cell differentiation accompanied by distinct microenvironmental alterations in preneoplastic breast tissues from BRCA1/2 mutation carriers and normal breast tissues from noncarrier controls. We uncovered spatially defined receptor-ligand interactions in these tissues for the investigation of autocrine and paracrine signaling. We discovered that β1-integrin-mediated autocrine signaling in BRCA2-deficient mammary epithelial cells may differ from BRCA1-deficient mammary epithelial cells. In addition, we found that the epithelial-to-stromal paracrine signaling in the breast tissues of BRCA1/2 mutation carriers is greater than in control tissues. More integrin-ligand pairs were differentially correlated in BRCA1/2-mutant breast tissues than noncarrier breast tissues with more integrin receptor-expressing stromal cells. IMPLICATIONS These results suggest alterations in the communication between mammary epithelial cells and the microenvironment in BRCA1 and BRCA2 mutation carriers, laying the foundation for designing innovative breast cancer chemo-prevention strategies for high-risk patients.
Collapse
Affiliation(s)
- Anthony Caputo
- Genomic Medicine Institute, Lerner Research Institute, Cleveland Clinic, Cleveland, OH, USA
| | - Kavya Vipparthi
- Genomic Medicine Institute, Lerner Research Institute, Cleveland Clinic, Cleveland, OH, USA
| | - Peter Bazeley
- Department of Quantitative Health Sciences, Lerner Research Institute, Cleveland Clinic, Cleveland, OH, USA
| | - Erinn Downs-Kelly
- Department of Pathology, Pathology & Laboratory Medicine Institute, Cleveland Clinic, Cleveland, OH, USA
| | - Patrick McIntire
- Department of Pathology, Pathology & Laboratory Medicine Institute, Cleveland Clinic, Cleveland, OH, USA
| | - Lauren A. Duckworth
- Department of Pathology, Pathology & Laboratory Medicine Institute, Cleveland Clinic, Cleveland, OH, USA
| | - Ying Ni
- Center for Immunotherapy & Precision Immuno-Oncology, Lerner Research Institute, Cleveland Clinic, Cleveland, OH, USA
| | - Bo Hu
- Department of Quantitative Health Sciences, Lerner Research Institute, Cleveland Clinic, Cleveland, OH, USA
| | - Ruth A. Keri
- Department of Cancer Biology, Lerner Research Institute, Cleveland Clinic, Cleveland, OH, USA
- Case Comprehensive Cancer Center, Case Western Reserve University School of Medicine, Cleveland, OH, USA
- Department of Molecular Medicine, Cleveland Clinic Lerner College of Medicine, Case Western Reserve University, Cleveland, OH, USA
| | - Mihriban Karaayvaz
- Genomic Medicine Institute, Lerner Research Institute, Cleveland Clinic, Cleveland, OH, USA
- Case Comprehensive Cancer Center, Case Western Reserve University School of Medicine, Cleveland, OH, USA
- Department of Molecular Medicine, Cleveland Clinic Lerner College of Medicine, Case Western Reserve University, Cleveland, OH, USA
| |
Collapse
|
190
|
Lone JB, Long JZ, Svensson KJ. Size matters: the biochemical logic of ligand type in endocrine crosstalk. LIFE METABOLISM 2024; 3:load048. [PMID: 38425548 PMCID: PMC10904031 DOI: 10.1093/lifemeta/load048] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/07/2023] [Revised: 11/21/2023] [Accepted: 12/04/2023] [Indexed: 03/02/2024]
Abstract
The endocrine system is a fundamental type of long-range cell-cell communication that is important for maintaining metabolism, physiology, and other aspects of organismal homeostasis. Endocrine signaling is mediated by diverse blood-borne ligands, also called hormones, including metabolites, lipids, steroids, peptides, and proteins. The size and structure of these hormones are fine-tuned to make them bioactive, responsive, and adaptable to meet the demands of changing environments. Why has nature selected such diverse ligand types to mediate communication in the endocrine system? What is the chemical, signaling, or physiologic logic of these ligands? What fundamental principles from our knowledge of endocrine communication can be applied as we continue as a field to uncover additional new circulating molecules that are claimed to mediate long-range cell and tissue crosstalk? This review provides a framework based on the biochemical logic behind this crosstalk with respect to their chemistry, temporal regulation in physiology, specificity, signaling actions, and evolutionary development.
Collapse
Affiliation(s)
- Jameel Barkat Lone
- Department of Pathology, Stanford University School of Medicine, Stanford, CA 94305, United States
| | - Jonathan Z Long
- Department of Pathology, Stanford University School of Medicine, Stanford, CA 94305, United States
- Department of Chemistry, Stanford University, Stanford, CA 94305, United States
- Stanford Diabetes Research Center, Stanford University School of Medicine, Stanford, CA 94305, United States
- Sarafan ChEM-H, Stanford University, Stanford, CA 94305, United States
- Wu Tsai Human Performance Alliance, Stanford University, Stanford, CA 94305, United States
| | - Katrin J Svensson
- Department of Pathology, Stanford University School of Medicine, Stanford, CA 94305, United States
- Stanford Diabetes Research Center, Stanford University School of Medicine, Stanford, CA 94305, United States
- Sarafan ChEM-H, Stanford University, Stanford, CA 94305, United States
- Wu Tsai Human Performance Alliance, Stanford University, Stanford, CA 94305, United States
- Stanford Cardiovascular Institute, Stanford University School of Medicine, CA 94305, United States
| |
Collapse
|
191
|
Wang M, Gao Z, Zhang Y, Zhao Q, Tan X, Wu S, Ding L, Liu Y, Qin S, Gu J, Xu L. Syringic acid promotes cartilage extracellular matrix generation and attenuates osteoarthritic cartilage degradation by activating TGF-β/Smad and inhibiting NF-κB signaling pathway. Phytother Res 2024; 38:1000-1012. [PMID: 38126609 DOI: 10.1002/ptr.8089] [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: 07/06/2023] [Revised: 11/14/2023] [Accepted: 11/26/2023] [Indexed: 12/23/2023]
Abstract
Osteoarthritis (OA) is a common chronic degenerative disease which is characterized by the disruption of articular cartilage. Syringic acid (SA) is a phenolic compound with anti-inflammatory, antioxidant, and other effects including promoting osteogenesis. However, the effect of SA on OA has not yet been reported. Therefore, the purpose of our study was to investigate the effect and mechanism of SA on OA in a mouse model of medial meniscal destabilization. The expressions of genes were evaluated by qPCR or western blot or immunofluorescence. RNA-seq analysis was performed to examine gene transcription alterations in chondrocytes treated with SA. The effect of SA on OA was evaluated using destabilization of the medial meniscus model of mice. We found that SA had no obvious toxic effect on chondrocytes, while promoting the expressions of chondrogenesis-related marker genes. The results of RNA-seq analysis showed that extracellular matrix-receptor interaction and transforming growth factor-β (TGF-β) signaling pathways were enriched among the up-regulated genes by SA. Mechanistically, we demonstrated that SA transcriptionally activated Smad3. In addition, we found that SA inhibited the overproduction of lipopolysaccharide-induced inflammation-related cytokines including tumor necrosis factor-α and interleukin-1β, as well as matrix metalloproteinase 3 and matrix metalloproteinase 13. The cell apoptosis and nuclear factor-kappa B (NF-κB) signaling were also inhibited by SA treatment. Most importantly, SA attenuated cartilage degradation in a mouse OA model. Taken together, our study demonstrated that SA could alleviate cartilage degradation in OA by activating the TGF-β/Smad and inhibiting NF-κB signaling pathway.
Collapse
Affiliation(s)
- Min Wang
- Key Laboratory of Orthopaedics and Traumatology, Lingnan Medical Research Center, The First Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Zhao Gao
- Er Sha Sports Training Center of Guangdong Province, Guangzhou, China
| | - Yage Zhang
- Key Laboratory of Orthopaedics and Traumatology, Lingnan Medical Research Center, The First Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Qiangqiang Zhao
- Key Laboratory of Orthopaedics and Traumatology, Lingnan Medical Research Center, The First Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Xinfang Tan
- Key Laboratory of Orthopaedics and Traumatology, Lingnan Medical Research Center, The First Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Siluo Wu
- Key Laboratory of Orthopaedics and Traumatology, Lingnan Medical Research Center, The First Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Lingli Ding
- Key Laboratory of Orthopaedics and Traumatology, Lingnan Medical Research Center, The First Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Yamei Liu
- School of Basic Medical Science, Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Shengnan Qin
- School of Biomedical Science, The University of Western Australia, Perth, Western Australia, Australia
| | - Jiangyong Gu
- School of Basic Medical Science, Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Liangliang Xu
- Key Laboratory of Orthopaedics and Traumatology, Lingnan Medical Research Center, The First Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou University of Chinese Medicine, Guangzhou, China
| |
Collapse
|
192
|
Wahbi W, Awad S, Salo T, Al-Samadi A. Stroma modulation of radiation response in head and neck squamous cell carcinoma: Insights from zebrafish larvae xenografts. Exp Cell Res 2024; 435:113911. [PMID: 38182078 DOI: 10.1016/j.yexcr.2024.113911] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2023] [Revised: 12/28/2023] [Accepted: 01/01/2024] [Indexed: 01/07/2024]
Abstract
BACKGROUND The tumour microenvironment (TME) of head and neck squamous cell carcinoma (HNSCC) consists of different subtypes of cells that interact with the tumour or with each other. This study investigates the possibility of co-culturing HNSCC cells with different stroma cells in a zebrafish xenograft model, focusing on the effect of stroma cells on HNSCC growth and response to irradiation. MATERIAL AND METHOD HNSCC metastatic cell line HSC-3 was used along with five types of stroma cells: normal gingival fibroblasts (NOF), cancer associated fibroblasts (CAF), macrophages, CD4+ T cells, and human umbilical vein endothelial cells (HUVEC). The mixture of HSC-3 cells and each-stroma cell type-was injected into 2-day post-fertilization zebrafish embryos, and the effect of stroma cells on tumour growth was tested. The study also aimed to mimic the HNSCC tumour by injecting a mixture of HSC-3 cells, CAFs, macrophages, and HUVECs into zebrafish embryos and testing the effect of these stroma cells on the cancer cells' response to irradiation compared to HSC-3-only tumours. RESULTS CAFs had a significant inducement effect on tumour size, while HUVECs showed the opposite effect. The irradiated group of HSC-3-only tumour had a significantly smaller tumor cell area compared to the control, while the group with stroma cells and HSC-3 cells showed cancer cells being resistant to irradiation. CONCLUSION This is the first report of co-culturing cancer cells with several types of stroma cells using a zebrafish xenograft model. This study also highlighted the role of stroma cells in turning the cancer cells from radioresponsive to radioresistant.
Collapse
Affiliation(s)
- Wafa Wahbi
- Department of Oral and Maxillofacial Diseases, Clinicum, Faculty of Medicine, University of Helsinki, Biomedicum Helsinki 1, C223b, Haartmaninkatu 8, P.O. Box 63, Helsinki, 00014, Finland; Translational Immunology Research Program (TRIMM), Faculty of Medicine, University of Helsinki, Biomedicum Helsinki 1, Haartmaninkatu 8, P.O. Box 63, Helsinki, 00014, Finland
| | - Shady Awad
- Clinical Pathology Department, National Cancer Institute, Cairo University, Cairo, Egypt; Hematology Research Unit, Department of Hematology, University of Helsinki and Helsinki University Central Hospital Comprehensive Cancer Center, Helsinki, Finland
| | - Tuula Salo
- Department of Oral and Maxillofacial Diseases, Clinicum, Faculty of Medicine, University of Helsinki, Biomedicum Helsinki 1, C223b, Haartmaninkatu 8, P.O. Box 63, Helsinki, 00014, Finland; Translational Immunology Research Program (TRIMM), Faculty of Medicine, University of Helsinki, Biomedicum Helsinki 1, Haartmaninkatu 8, P.O. Box 63, Helsinki, 00014, Finland; Department of Pathology, HUSLAB, University of Helsinki and Helsinki University Hospital, P.O. Box 21, Helsinki, 00014, Finland; Cancer and Translational Medicine Research Unit, Faculty of Medicine, University of Oulu, P.O. Box 5281, Oulu, 90014, Finland; Medical Research Center Oulu, Oulu University Hospital and University of Oulu, P.O. Box 5281, Oulu, 90014, Finland
| | - Ahmed Al-Samadi
- Department of Oral and Maxillofacial Diseases, Clinicum, Faculty of Medicine, University of Helsinki, Biomedicum Helsinki 1, C223b, Haartmaninkatu 8, P.O. Box 63, Helsinki, 00014, Finland; Translational Immunology Research Program (TRIMM), Faculty of Medicine, University of Helsinki, Biomedicum Helsinki 1, Haartmaninkatu 8, P.O. Box 63, Helsinki, 00014, Finland; Institute of Dentistry, School of Medicine, Kuopio Campus, University of Eastern Finland, P.O. Box 1627, Kuopio, Finland.
| |
Collapse
|
193
|
Tseng C, Chen B, Han Y, Wang K, Song Q, Shen H, Chen Z. Advanced glycation end products promote intervertebral disc degeneration by transactivation of matrix metallopeptidase genes. Osteoarthritis Cartilage 2024; 32:187-199. [PMID: 37717904 DOI: 10.1016/j.joca.2023.09.005] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/17/2023] [Revised: 09/05/2023] [Accepted: 09/11/2023] [Indexed: 09/19/2023]
Abstract
OBJECTIVE Examine the mechanism by which advanced glycation end products (AGEs) induce intervertebral disc degeneration (IDD) in C57BL/6J mice. METHODS Matrix metallopeptidase (MMP) gene mRNA levels were assessed using RT-qPCR. Immunoprecipitation and co-immunoprecipitation were performed to identify the transcriptional complex regulating MMP expression due to AGEs. The preventive effects of inhibitors targeting this complex were tested in mice on high AGE diets. RESULTS IDD and AGE accumulation were evident in mice on high-AGE diets (HAGEs), persisting across dietary shifts but absent in mice exclusively on low-AGE diets. Molecularly, HAGEs activated p21-activated kinase 1 (PAK1), prompting peroxisome proliferator-activated receptor gamma coactivator-related protein 1 (PPRC1) phosphorylation. Ubiquitin-specific protease 12 (USP12) interacted with the phosphorylated PPRC1 (pPPRC1), safeguarding it from proteasomal degradation. This pPPRC1, in collaboration with two histone acetyltransferases p300/CREB-binding protein (CBP) and a transcription factor activator protein 1(AP1), enhanced the expression of 12 MMP genes (MMP1a/1b/3/7/9/10/12/13/16/19/23/28). In vitro AGE exposure on nucleus pulposus and annulus fibrosus cells replicated this gene activation pattern, driven by the PAK1/pPPRC1-p300/CBP-AP1 pathway. The application of PAK1, p300, and AP1 inhibitors reduced pPPRC1-p300/CBP-AP1 binding to MMP promoters, diminishing their expression. These inhibitors effectively thwarted IDD in HAGE mice. CONCLUSION Our results revealed that HAGEs instigate IDD via the PAK1/pPPRC1-p300/CBP-AP1 signaling pathway. This insight can guide therapeutic strategies to slow IDD progression in prediabetic/diabetic patients.
Collapse
Affiliation(s)
- Changchun Tseng
- Department of Spine Surgery, Department of Orthopedics, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Bin Chen
- Department of Spine Surgery, Department of Orthopedics, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Yingchao Han
- Department of Spine Surgery, Department of Orthopedics, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Kun Wang
- Department of Spine Surgery, Department of Orthopedics, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Qingxin Song
- Department of Spine Surgery, Department of Orthopedics, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Hongxing Shen
- Department of Spine Surgery, Department of Orthopedics, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Zhi Chen
- Department of Spine Surgery, Department of Orthopedics, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China.
| |
Collapse
|
194
|
Yang F, Hu Y, Shi Z, Liu M, Hu K, Ye G, Pang Q, Hou R, Tang K, Zhu Y. The occurrence and development mechanisms of esophageal stricture: state of the art review. J Transl Med 2024; 22:123. [PMID: 38297325 PMCID: PMC10832115 DOI: 10.1186/s12967-024-04932-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2023] [Accepted: 01/26/2024] [Indexed: 02/02/2024] Open
Abstract
BACKGROUND Esophageal strictures significantly impair patient quality of life and present a therapeutic challenge, particularly due to the high recurrence post-ESD/EMR. Current treatments manage symptoms rather than addressing the disease's etiology. This review concentrates on the mechanisms of esophageal stricture formation and recurrence, seeking to highlight areas for potential therapeutic intervention. METHODS A literature search was conducted through PUBMED using search terms: esophageal stricture, mucosal resection, submucosal dissection. Relevant articles were identified through manual review with reference lists reviewed for additional articles. RESULTS Preclinical studies and data from animal studies suggest that the mechanisms that may lead to esophageal stricture include overdifferentiation of fibroblasts, inflammatory response that is not healed in time, impaired epithelial barrier function, and multimethod factors leading to it. Dysfunction of the epithelial barrier may be the initiating mechanism for esophageal stricture. Achieving perfect in-epithelialization by tissue-engineered fabrication of cell patches has been shown to be effective in the treatment and prevention of esophageal strictures. CONCLUSION The development of esophageal stricture involves three stages: structural damage to the esophageal epithelial barrier (EEB), chronic inflammation, and severe fibrosis, in which dysfunction or damage to the EEB is the initiating mechanism leading to esophageal stricture. Re-epithelialization is essential for the treatment and prevention of esophageal stricture. This information will help clinicians or scientists to develop effective techniques to treat esophageal stricture in the future.
Collapse
Affiliation(s)
- Fang Yang
- Health Science Center, Ningbo University, Ningbo, 315211, People's Republic of China
| | - Yiwei Hu
- Health Science Center, Ningbo University, Ningbo, 315211, People's Republic of China
| | - Zewen Shi
- Health Science Center, Ningbo University, Ningbo, 315211, People's Republic of China
- Ningbo No.2 Hospital, Ningbo, 315001, People's Republic of China
| | - Mujie Liu
- Health Science Center, Ningbo University, Ningbo, 315211, People's Republic of China
| | - Kefeng Hu
- The First Affiliated Hospital of Ningbo University, Ningbo, 315000, People's Republic of China
| | - Guoliang Ye
- The First Affiliated Hospital of Ningbo University, Ningbo, 315000, People's Republic of China
| | - Qian Pang
- Health Science Center, Ningbo University, Ningbo, 315211, People's Republic of China
| | - Ruixia Hou
- Health Science Center, Ningbo University, Ningbo, 315211, People's Republic of China
| | - Keqi Tang
- Institute of Mass Spectrometry, School of Material Science and Chemical Engineering, Ningbo University, Ningbo, 315211, People's Republic of China.
| | - Yabin Zhu
- Health Science Center, Ningbo University, Ningbo, 315211, People's Republic of China.
| |
Collapse
|
195
|
Du W, Xia X, Hu F, Yu J. Extracellular matrix remodeling in the tumor immunity. Front Immunol 2024; 14:1340634. [PMID: 38332915 PMCID: PMC10850336 DOI: 10.3389/fimmu.2023.1340634] [Citation(s) in RCA: 24] [Impact Index Per Article: 24.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2023] [Accepted: 12/28/2023] [Indexed: 02/10/2024] Open
Abstract
The extracellular matrix (ECM) is a significant constituent of tumors, fulfilling various essential functions such as providing mechanical support, influencing the microenvironment, and serving as a reservoir for signaling molecules. The abundance and degree of cross-linking of ECM components are critical determinants of tissue stiffness. In the process of tumorigenesis, the interaction between ECM and immune cells within the tumor microenvironment (TME) frequently leads to ECM stiffness, thereby disrupting normal mechanotransduction and promoting malignant progression. Therefore, acquiring a thorough comprehension of the dysregulation of ECM within the TME would significantly aid in the identification of potential therapeutic targets for cancer treatment. In this regard, we have compiled a comprehensive summary encompassing the following aspects: (1) the principal components of ECM and their roles in malignant conditions; (2) the intricate interaction between ECM and immune cells within the TME; and (3) the pivotal regulators governing the onco-immune response in ECM.
Collapse
Affiliation(s)
- Wei Du
- Department of Targeting Therapy and Immunology, West China Hospital, Sichuan University, Chengdu, China
| | - Xueming Xia
- Division of Head & Neck Tumor Multimodality Treatment, Cancer Center, West China Hospital, Sichuan University, Chengdu, China
| | - Fan Hu
- Key Laboratory of Birth Defects and Related Diseases of Women and Children, West China Second University Hospital, Sichuan University, Chengdu, China
| | - Jiayun Yu
- Department of Radiation Oncology, Cancer Center, West China Hospital, Sichuan University, Chengdu, China
| |
Collapse
|
196
|
Müller P, Zimmer C, Frey A, Holzmann G, Weldert AC, Schirmeister T. Ligand-Based Design of Selective Peptidomimetic uPA and TMPRSS2 Inhibitors with Arg Bioisosteres. Int J Mol Sci 2024; 25:1375. [PMID: 38338655 PMCID: PMC10855164 DOI: 10.3390/ijms25031375] [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: 01/03/2024] [Revised: 01/20/2024] [Accepted: 01/21/2024] [Indexed: 02/12/2024] Open
Abstract
Trypsin-like serine proteases are involved in many important physiological processes like blood coagulation and remodeling of the extracellular matrix. On the other hand, they are also associated with pathological conditions. The urokinase-pwlasminogen activator (uPA), which is involved in tissue remodeling, can increase the metastatic behavior of various cancer types when overexpressed and dysregulated. Another member of this protease class that received attention during the SARS-CoV 2 pandemic is TMPRSS2. It is a transmembrane serine protease, which enables cell entry of the coronavirus by processing its spike protein. A variety of different inhibitors have been published against both proteases. However, the selectivity over other trypsin-like serine proteases remains a major challenge. In the current study, we replaced the arginine moiety at the P1 site of peptidomimetic inhibitors with different bioisosteres. Enzyme inhibition studies revealed that the phenylguanidine moiety in the P1 site led to strong affinity for TMPRSS2, whereas the cyclohexylguanidine derivate potently inhibited uPA. Both inhibitors exhibited high selectivity over other structurally similar and physiologically important proteases.
Collapse
Affiliation(s)
| | | | | | | | | | - Tanja Schirmeister
- Institute of Pharmaceutical and Biomedical Sciences, Johannes Gutenberg University Mainz, Staudinger Weg 5, D-55128 Mainz, Germany; (P.M.); (C.Z.); (A.F.); (G.H.); (A.C.W.)
| |
Collapse
|
197
|
Huang X, Zhang L, Luo W, Zeng Y, Li X, Yang N, Huang W, Ding BS. Endothelial anthrax toxin receptor 2 plays a protective role in liver fibrosis. Front Cell Dev Biol 2024; 11:1278968. [PMID: 38322497 PMCID: PMC10844529 DOI: 10.3389/fcell.2023.1278968] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2023] [Accepted: 12/18/2023] [Indexed: 02/08/2024] Open
Abstract
Hepatocellular carcinoma is one of the leading cancers worldwide and is a potential consequence of fibrosis. Therefore, the identification of key cellular and molecular mechanisms involved in liver fibrosis is an important goal for the development of new strategies to control liver-related diseases. Here, single-cell RNA sequencing data (GSE136103 and GES181483) of clinical liver non-parenchymal cells were analyzed to identify cellular and molecular mechanisms of liver fibrosis. The proportion of endothelial subpopulations in cirrhotic livers was significantly higher than that in healthy livers. Gene ontology and gene set enrichment analysis of differentially expressed genes in the endothelial subgroups revealed that extracellular matrix (ECM)-related pathways were significantly enriched. Since anthrax toxin receptor 2 (ANTXR2) interacts with the ECM, the expression of ANTXR2 in the liver endothelium was analyzed. ANTXR2 expression in the liver endothelium of wild-type (WT) mice significantly decreased after a 4-time sequential injection of carbon tetrachloride (CCl4) to induce liver fibrosis. Next, conditional knockout mice selectively lacking Antxr2 in endothelial cells were generated. After endothelial-specific Antxr2 knockout mice were subjected to the CCl4 model, the degree of liver fibrosis in the knockout group was significantly more severe than that in the control group. In addition, ANTXR2 in human umbilical vein endothelial cells promoted matrix metalloproteinase 2 (MMP2) activation to degrade the ECM in vitro. Finally, endothelial-specific overexpression of Antxr2 alleviated the development of liver fibrosis following adeno-associated virus treatment. Collectively, these results suggested that endothelial ANTXR2 plays a protective role in liver fibrosis. This function of ANTXR2 may be achieved by promoting MMP2 activation to degrade the ECM.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | - Bi-Sen Ding
- Key Laboratory of Birth Defects and Related Diseases of Women and Children of MOE, State Key Laboratory of Biotherapy, West China Second University Hospital, Sichuan University, Chengdu, China
| |
Collapse
|
198
|
Didwischus N, Guduru A, Badylak SF, Modo M. In vitro dose-dependent effects of matrix metalloproteinases on ECM hydrogel biodegradation. Acta Biomater 2024; 174:104-115. [PMID: 38081445 PMCID: PMC10775082 DOI: 10.1016/j.actbio.2023.12.003] [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: 07/17/2023] [Revised: 11/17/2023] [Accepted: 12/04/2023] [Indexed: 12/18/2023]
Abstract
Matrix metalloproteinases (MMPs) cause proteolysis of extracellular matrix (ECM) in tissues affected by stroke. However, little is known about how MMPs degrade ECM hydrogels implanted into stroke cavities to regenerate lost tissue. To establish a structure-function relationship between different doses of individual MMPs and isolate their effects in a controlled setting, an in vitro degradation assay quantified retained urinary bladder matrix (UBM) hydrogel mass as a measure of degradation across time. A rheological characterization indicated that lower ECM concentrations (<4 mg/mL) did not cure completely at 37 °C and had a high fraction of mobile proteins that were easily washed-out. Hydrolysis by dH2O caused a steady 2 % daily decrease in hydrogel mass over 14 days. An acceleration of degradation to 6 % occurred with phosphate buffered saline and artificial cerebrospinal fluid. MMPs induced a dose-dependent increase and within 14 days almost completely (>95 %) degraded the hydrogel. MMP-9 exerted the most significant biodegradation, compared to MMP-3 and -2. To model the in vivo exposure of hydrogel to MMPs, mixtures of MMP-2, -3, and -9, present in the cavity at 14-, 28-, or 90-days post-stroke, revealed that 14- and 28-days mixtures achieved an equivalent biodegradation, but a 90-days mixture exhibited a slower degradation. These results revealed that hydrolysis, in addition to proteolysis, exerts a major influence on the degradation of hydrogels. Understanding the mechanisms of ECM hydrogel biodegradation is essential to determine the therapeutic window for bioscaffold implantation after a stroke, and they are also key to determine optimal degradation kinetics to support tissue regeneration. STATEMENT OF SIGNIFICANCE: After implantation into a stroke cavity, extracellular matrix (ECM) hydrogel promotes tissue regeneration through the degradation of the bioscaffold. However, the process of degradation of an ECM hydrogel remains poorly understood. We here demonstrated in vitro under highly controlled conditions that hydrogel degradation is very dependent on its protein concentration. Lower protein concentration hydrogels were weaker in rheological measurements and particularly susceptible to hydrolysis. The proteolytic degradation of tissue ECM after a stroke is caused by matrix metalloproteinases (MMPs). A dose-dependent MMP-driven biodegradation of ECM hydrogel exceeded the effects of hydrolysis. These results highlight the importance of in vitro testing of putative causes of degradation to gain a better understanding of how these factors affect in vivo biodegradation.
Collapse
Affiliation(s)
- Nadine Didwischus
- Department of Radiology, University of Pittsburgh, Pittsburgh, PA, USA; McGowan Institute for Regenerative Medicine, University of Pittsburgh, Pittsburgh, PA, USA
| | - Arun Guduru
- Department of Bioengineering, University of Pittsburgh, Pittsburgh, PA, USA
| | - Stephen F Badylak
- McGowan Institute for Regenerative Medicine, University of Pittsburgh, Pittsburgh, PA, USA; Department of Bioengineering, University of Pittsburgh, Pittsburgh, PA, USA; Department of Surgery, University of Pittsburgh, Pittsburgh, PA, USA
| | - Michel Modo
- Department of Radiology, University of Pittsburgh, Pittsburgh, PA, USA; McGowan Institute for Regenerative Medicine, University of Pittsburgh, Pittsburgh, PA, USA; Department of Bioengineering, University of Pittsburgh, Pittsburgh, PA, USA.
| |
Collapse
|
199
|
Rasti Boroojeni F, Naeimipour S, Lifwergren P, Abrahamsson A, Dabrosin C, Selegård R, Aili D. Proteolytic remodeling of 3D bioprinted tumor microenvironments. Biofabrication 2024; 16:025002. [PMID: 38128125 DOI: 10.1088/1758-5090/ad17d1] [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: 09/13/2023] [Accepted: 12/21/2023] [Indexed: 12/23/2023]
Abstract
In native tissue, remodeling of the pericellular space is essential for cellular activities and is mediated by tightly regulated proteases. Protease activity is dysregulated in many diseases, including many forms of cancer. Increased proteolytic activity is directly linked to tumor invasion into stroma, metastasis, and angiogenesis as well as all other hallmarks of cancer. Here we show a strategy for 3D bioprinting of breast cancer models using well-defined protease degradable hydrogels that can facilitate exploration of the multifaceted roles of proteolytic extracellular matrix remodeling in tumor progression. We designed a set of bicyclo[6.1.0]nonyne functionalized hyaluronan (HA)-based bioinks cross-linked by azide-modified poly(ethylene glycol) (PEG) or matrix metalloproteinase (MMP) degradable azide-functionalized peptides. Bioprinted structures combining PEG and peptide-based hydrogels were proteolytically degraded with spatial selectivity, leaving non-degradable features intact. Bioprinting of tumor-mimicking microenvironments using bioinks comprising human breast cancer cells (MCF-7) and fibroblast in hydrogels with different susceptibilities to proteolytic degradation shows that MCF-7 proliferation and spheroid size were significantly increased in protease degradable hydrogel compartments, but only in the presence of fibroblasts. In the absence of fibroblasts in the stromal compartment, cancer cell proliferation was reduced and did not differ between degradable and nondegradable hydrogels. The interactions between spatially separated fibroblasts and MCF-7 cells consequently resulted in protease-mediated remodeling of the bioprinted structures and a significant increase in cancer cell spheroid size, highlighting the close interplay between cancer cells and stromal cells in the tumor microenvironment and the influence of proteases in tumor progression.
Collapse
Affiliation(s)
- Fatemeh Rasti Boroojeni
- Laboratory of Molecular Materials, Division of Biophysics and Bioengineering, Department of Physics, Chemistry and Biology, Linköping University, 581 83 Linköping, Sweden
| | - Sajjad Naeimipour
- Laboratory of Molecular Materials, Division of Biophysics and Bioengineering, Department of Physics, Chemistry and Biology, Linköping University, 581 83 Linköping, Sweden
| | - Philip Lifwergren
- Laboratory of Molecular Materials, Division of Biophysics and Bioengineering, Department of Physics, Chemistry and Biology, Linköping University, 581 83 Linköping, Sweden
| | - Annelie Abrahamsson
- Department of Oncology and Department of Biomedical and Clinical Sciences, Linköping University, Linköping, Sweden
| | - Charlotta Dabrosin
- Department of Oncology and Department of Biomedical and Clinical Sciences, Linköping University, Linköping, Sweden
| | - Robert Selegård
- Laboratory of Molecular Materials, Division of Biophysics and Bioengineering, Department of Physics, Chemistry and Biology, Linköping University, 581 83 Linköping, Sweden
| | - Daniel Aili
- Laboratory of Molecular Materials, Division of Biophysics and Bioengineering, Department of Physics, Chemistry and Biology, Linköping University, 581 83 Linköping, Sweden
| |
Collapse
|
200
|
Costa D, Scalise E, Ielapi N, Bracale UM, Andreucci M, Serra R. Metalloproteinases as Biomarkers and Sociomarkers in Human Health and Disease. Biomolecules 2024; 14:96. [PMID: 38254696 PMCID: PMC10813678 DOI: 10.3390/biom14010096] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2023] [Revised: 01/05/2024] [Accepted: 01/10/2024] [Indexed: 01/24/2024] Open
Abstract
Metalloproteinases (MPs) are zinc-dependent enzymes with proteolytic activity and a variety of functions in the pathophysiology of human diseases. The main objectives of this review are to analyze a specific family of MPs, the matrix metalloproteinases (MMPs), in the most common chronic and complex diseases that affect patients' social lives and to better understand the nature of the associations between MMPs and the psychosocial environment. In accordance with the PRISMA extension for a scoping review, an examination was carried out. A collection of 24 studies was analyzed, focusing on the molecular mechanisms of MMP and their connection to the manifestation of social aspects in human disease. The complexity of the relationship between MMP and social problems is presented via an interdisciplinary approach based on complexity paradigm as a new approach for conceptualizing knowledge in health research. Finally, two implications emerge from the study: first, the psychosocial states of individuals have a profound impact on their overall health and disease conditions, which implies the importance of adopting a holistic perspective on human well-being, encompassing both physical and psychosocial aspects. Second, the use of MPs as biomarkers may provide physicians with valuable tools for a better understanding of disease when used in conjunction with "sociomarkers" to develop mathematical predictive models.
Collapse
Affiliation(s)
- Davide Costa
- Department of Medical and Surgical Sciences, Magna Graecia University of Catanzaro, 88100 Catanzaro, Italy; (D.C.); (E.S.)
- Interuniversity Center of Phlebolymphology (CIFL), Magna Graecia University of Catanzaro, 88100 Catanzaro, Italy
| | - Enrica Scalise
- Department of Medical and Surgical Sciences, Magna Graecia University of Catanzaro, 88100 Catanzaro, Italy; (D.C.); (E.S.)
- Interuniversity Center of Phlebolymphology (CIFL), Magna Graecia University of Catanzaro, 88100 Catanzaro, Italy
| | - Nicola Ielapi
- Department of Public Health and Infectious Disease, “Sapienza” University of Rome, 00185 Rome, Italy;
| | | | - Michele Andreucci
- Department of Health Sciences, Magna Graecia University of Catanzaro, 88100 Catanzaro, Italy
| | - Raffaele Serra
- Department of Medical and Surgical Sciences, Magna Graecia University of Catanzaro, 88100 Catanzaro, Italy; (D.C.); (E.S.)
- Interuniversity Center of Phlebolymphology (CIFL), Magna Graecia University of Catanzaro, 88100 Catanzaro, Italy
| |
Collapse
|