1
|
Gunter NV, Ong YS, Lai ZW, Morita H, Chamyuang S, Owatworakit A, Mah SH. Anti-cancer effects of cordyceps sinensis, C. militaris and C. cicadae and their mechanisms of action. JOURNAL OF ASIAN NATURAL PRODUCTS RESEARCH 2025:1-25. [PMID: 40372357 DOI: 10.1080/10286020.2025.2497282] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/01/2024] [Revised: 04/15/2025] [Accepted: 04/15/2025] [Indexed: 05/16/2025]
Abstract
Over the past decade, species of Cordyceps, particularly C. sinensis, C. militaris, and C. cicadae, along with their bioactive components cordycepin, ergosterol and polysaccharides, have shown significant anti-cancer potential. These effects are mediated through mechanisms including apoptosis induction, cell cycle arrest, and modulation of key signalling pathways, as reviewed here. Notably, the bioactive components exhibit greater potency than crude extracts, with cordycepin being a promising chemotherapeutic lead. Future investigations into the in vivo efficacy and safety profiles of Cordyceps extracts and their bioactive components are greatly encouraged. Additionally, structural modification of cordycepin may offer opportunities to enhance its therapeutic potential.
Collapse
Affiliation(s)
- Natalie Vivien Gunter
- School of Biosciences, Taylor's University, Lakeside Campus, Selangor, 47500, Malaysia
| | - Yee Swen Ong
- School of Biosciences, Taylor's University, Lakeside Campus, Selangor, 47500, Malaysia
| | - Zee Wei Lai
- School of Biosciences, Taylor's University, Lakeside Campus, Selangor, 47500, Malaysia
| | - Hiroyuki Morita
- Institute of Natural Medicine, University of Toyama, Toyama, 930-0194, Japan
| | - Sunita Chamyuang
- School of Science, Mae Fah Luang University, Chiang Rai, 57100, Thailand
- Microbial Products and Innovation Research Group, School of Science, Mae Fah Luang University, Chiang Rai, 57100, Thailand
| | - Amorn Owatworakit
- School of Science, Mae Fah Luang University, Chiang Rai, 57100, Thailand
- Microbial Products and Innovation Research Group, School of Science, Mae Fah Luang University, Chiang Rai, 57100, Thailand
| | - Siau Hui Mah
- School of Biosciences, Taylor's University, Lakeside Campus, Selangor, 47500, Malaysia
| |
Collapse
|
2
|
Roamcharern N, Matthew SAL, Brady DJ, Parkinson JA, Rattray Z, Seib FP. Biomimetic Silk Nanoparticle Manufacture: Calcium Ion-Mediated Assembly. ACS Biomater Sci Eng 2025; 11:1847-1856. [PMID: 39883858 PMCID: PMC11897946 DOI: 10.1021/acsbiomaterials.4c02175] [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: 11/18/2024] [Revised: 01/08/2025] [Accepted: 01/15/2025] [Indexed: 02/01/2025]
Abstract
Silk has emerged as an interesting candidate among protein-based nanocarriers due to its favorable properties, including biocompatibility and a broad spectrum of processing options to tune particle critical quality attributes. The silk protein conformation during storage in the middle silk gland of the silkworm is modulated by various factors, including the most abundant metallic ion, calcium ion (Ca2+). Here, we report spiking of liquid silk with calcium ions to modulate the silk nanoparticle size. Conformational and structural analyses of silk demonstrated Ca2+-induced silk assemblies that resulted in a liquid crystalline-like state, with the subsequent generation of β-sheet-enriched silk nanoparticles. Thioflavin T studies demonstrated that Ca2+ effectively induces self-assembly and conformation changes that also increased model drug loading. Ca2+ incorporation in the biopolymer feed significantly increased the nanoparticle production yield from 16 to 89%, while simultaneously enabling Ca2+ concentration-dependent particle-size tuning with a narrow polydispersity index and altered zeta potential. The resulting silk nanoparticles displayed high biocompatibility in macrophages with baseline levels of cytotoxicity and cellular inflammation. Our strategy for manufacturing biomimetic silk nanoparticles enabled overall tuning of particle size and improved yields─features that are critical for particle-based nanomedicines.
Collapse
Affiliation(s)
- Napaporn Roamcharern
- Strathclyde
Institute of Pharmacy and Biomedical Sciences, University of Strathclyde, 161 Cathedral St., Glasgow G4 0RE,Scotland,U.K.
| | - Saphia A. L. Matthew
- Strathclyde
Institute of Pharmacy and Biomedical Sciences, University of Strathclyde, 161 Cathedral St., Glasgow G4 0RE,Scotland,U.K.
| | - Daniel J. Brady
- Branch Bioresources, Fraunhofer Institute for Molecular Biology and Applied
Ecology, Ohlebergsweg
12, Giessen 35392, Germany
| | - John A. Parkinson
- Department
of Pure and Applied Chemistry, University
of Strathclyde, 295 Cathedral Street, Glasgow G1 1XL, Scotland,U.K.
| | - Zahra Rattray
- Strathclyde
Institute of Pharmacy and Biomedical Sciences, University of Strathclyde, 161 Cathedral St., Glasgow G4 0RE,Scotland,U.K.
| | - F. Philipp Seib
- Strathclyde
Institute of Pharmacy and Biomedical Sciences, University of Strathclyde, 161 Cathedral St., Glasgow G4 0RE,Scotland,U.K.
- Branch Bioresources, Fraunhofer Institute for Molecular Biology and Applied
Ecology, Ohlebergsweg
12, Giessen 35392, Germany
- Institute
of Pharmacy, Department of Pharmaceutics and Biopharmaceutics, Friedrich Schiller University Jena, Lessingstr. 8, Jena 07743, Germany
| |
Collapse
|
3
|
Duzen IV, Tuluce SY, Ozturk S, Savcılıoglu MD, Goksuluk H, Altunbas G, Kaplan M, Vuruskan E, Tabur S, Sucu M, Taysi S. Assessment of Left Ventricular Strain Echocardiography in Individuals with Hashimoto's Thyroiditis and Its Association with Serum TIMP-1 Concentration. J Clin Med 2025; 14:1705. [PMID: 40095713 PMCID: PMC11900031 DOI: 10.3390/jcm14051705] [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: 01/18/2025] [Revised: 02/15/2025] [Accepted: 02/25/2025] [Indexed: 03/19/2025] Open
Abstract
Background: Hashimoto's thyroiditis (HT), which is an autoimmune condition and the primary cause of hypothyroidism, has numerous impacts on the cardiovascular system. This research aimed to compare TIMP-1 levels and LV strain values in euthyroid HT, hypothyroid HT, and healthy control persons. Materials and Methods: This study included 40 hypothyroid HT patients, 42 HT patients who became euthyroid with thyroid hormone replacement therapy, and 40 healthy controls. All subjects had conventional echocardiography and STE. Global and segmental LV longitudinal strain values (LVGLS) were calculated. Participants' blood was tested for TIMP-1, thyroid function, and anti-TPO. Results: Higher serum TIMP-1 levels were found in euthyroid and hypothyroid HT patients than in the control group. Additionally, patients with euthyroid and hypothyroid HT displayed lower segmental and global LV strain values than the control group. A negative correlation was observed between strain values and TIMP-1 and anti-TPO levels. No significant difference was observed in serum TIMP-1 and strain values between euthyroid and hypothyroid HT patients. Patients with hypothyroid HT exhibited impaired diastolic function and reduced ejection fraction when compared to both euthyroid HT and control groups. However, euthyroid HT patients and the controls had similar diastolic function and ejection fractions. Conclusions: Hashimoto's thyroiditis causes impairment of LV strain, regardless of thyroid hormone levels. Additionally, the condition is associated with elevated TIMP-1 levels. The relationship between LV strain values and anti-TPO levels indicates that the autoimmune component of the disease may be responsible for the impaired LV strain.
Collapse
Affiliation(s)
- Irfan V. Duzen
- Department of Cardiology, Faculty of Medicine, Gaziantep University Sahinbey Education and Research Hospital, Gaziantep University, Gaziantep 27310, Turkey; (G.A.); (M.K.); (E.V.); (M.S.)
| | - Selcen Y. Tuluce
- Department of Cardiology, Cardiology Clinic, Heart Izmir Clinic, Izmir 35610, Turkey;
| | - Sadettin Ozturk
- Department of Endocrinology and Metabolic Disease, Gaziantep City Hospital, Gaziantep 27470, Turkey;
| | - Mert D. Savcılıoglu
- Department of Cardiology, Cardiology Clinic, Gaziantep City Hospital, Gaziantep 27470, Turkey;
| | - Huseyin Goksuluk
- Department of Cardiology, Cardiology Clinic, Bursa Anadolu Hospital, Bursa 16320, Turkey;
| | - Gokhan Altunbas
- Department of Cardiology, Faculty of Medicine, Gaziantep University Sahinbey Education and Research Hospital, Gaziantep University, Gaziantep 27310, Turkey; (G.A.); (M.K.); (E.V.); (M.S.)
| | - Mehmet Kaplan
- Department of Cardiology, Faculty of Medicine, Gaziantep University Sahinbey Education and Research Hospital, Gaziantep University, Gaziantep 27310, Turkey; (G.A.); (M.K.); (E.V.); (M.S.)
| | - Ertan Vuruskan
- Department of Cardiology, Faculty of Medicine, Gaziantep University Sahinbey Education and Research Hospital, Gaziantep University, Gaziantep 27310, Turkey; (G.A.); (M.K.); (E.V.); (M.S.)
| | - Suzan Tabur
- Department of Endocrinology and Metabolic Disease, Faculty of Medicine, Gaziantep University Sahinbey Education and Research Hospital, Gaziantep University, Gaziantep 27310, Turkey;
| | - Murat Sucu
- Department of Cardiology, Faculty of Medicine, Gaziantep University Sahinbey Education and Research Hospital, Gaziantep University, Gaziantep 27310, Turkey; (G.A.); (M.K.); (E.V.); (M.S.)
| | - Seyithan Taysi
- Department of Medical Biochemistry, Faculty of Medicine, Gaziantep University Sahinbey Education and Research Hospital, Gaziantep University, Gaziantep 27310, Turkey;
| |
Collapse
|
4
|
Xu J, Li Y, Feng Z, Chen H. Cigarette Smoke Contributes to the Progression of MASLD: From the Molecular Mechanisms to Therapy. Cells 2025; 14:221. [PMID: 39937012 PMCID: PMC11816580 DOI: 10.3390/cells14030221] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2024] [Revised: 01/22/2025] [Accepted: 01/31/2025] [Indexed: 02/13/2025] Open
Abstract
Cigarette smoke (CS), an intricate blend comprising over 4000 compounds, induces abnormal cellular reactions that harm multiple tissues. Non-alcoholic fatty liver disease (NAFLD) is a prevalent chronic liver disease (CLD), encompassing non-alcoholic fatty liver (NAFL), non-alcoholic steatohepatitis (NASH), cirrhosis, and hepatocellular carcinoma (HCC). Recently, the term NAFLD has been changed to metabolic dysfunction-associated steatotic liver disease (MASLD), and NASH has been renamed metabolic dysfunction-associated steatohepatitis (MASH). A multitude of experiments have confirmed the association between CS and the incidence and progression of MASLD. However, the specific signaling pathways involved need to be updated with new scientific discoveries. CS exposure can disrupt lipid metabolism, induce inflammation and apoptosis, and stimulate liver fibrosis through multiple signaling pathways that promote the progression of MASLD. Currently, there is no officially approved efficacious pharmaceutical intervention in clinical practice. Therefore, lifestyle modifications have emerged as the primary therapeutic approach for managing MASLD. Smoking cessation and the application of a series of natural ingredients have been shown to ameliorate pathological changes in the liver induced by CS, potentially serving as an effective approach to decelerating MASLD development. This article aims to elucidate the specific signaling pathways through which smoking promotes MASLD, while summarizing the reversal factors identified in recent studies, thereby offering novel insights for future research on and the treatment of MASLD.
Collapse
Affiliation(s)
- Jiatong Xu
- Queen Mary School, Medical College, Nanchang University, Nanchang 330006, China; (J.X.); (Y.L.); (Z.F.)
| | - Yifan Li
- Queen Mary School, Medical College, Nanchang University, Nanchang 330006, China; (J.X.); (Y.L.); (Z.F.)
| | - Zixuan Feng
- Queen Mary School, Medical College, Nanchang University, Nanchang 330006, China; (J.X.); (Y.L.); (Z.F.)
| | - Hongping Chen
- Department of Histology and Embryology, Jiangxi Medical College, Nanchang University, Nanchang 330019, China
| |
Collapse
|
5
|
Rieber J, Niederhauser RK, Giovanoli P, Buschmann J. Fabrication and Characterization of Electrospun DegraPol ® Tubes Releasing TIMP-1 Protein to Modulate Tendon Healing. MATERIALS (BASEL, SWITZERLAND) 2025; 18:665. [PMID: 39942332 PMCID: PMC11820012 DOI: 10.3390/ma18030665] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 12/20/2024] [Revised: 01/15/2025] [Accepted: 01/30/2025] [Indexed: 02/16/2025]
Abstract
BACKGROUND Tendon rupture repair can result from fibrotic scar formation through imbalanced ECM deposition during remodeling. The tissue inhibitors of matrix metalloprotease (TIMPs) not only decrease ECM degradation, regulated by matrix metalloproteases (MMPs), but also restrict TGF-β1 activation and thus diminish fibrosis. METHODS Rabbit tenocytes (rbTenocytes) and rabbit adipose-derived stem cells (rbASCs) were cultivated under different TIMP-1 concentrations. Proliferation and gene expression were assessed. TIMP-1 was incorporated into emulsion electrospun DegraPol® (DP) tubes that were characterized by SEM for fiber thickness, pore size, and wall thickness. Static and dynamic water contact angles, FTIR spectra, and TIMP-1 release kinetics were determined. RESULTS While the proliferation of rbTenocytes and rbACS was not affected by TIMP-1 supplementation in vitro, the gene expression of Col1A1 was increased in rbTenocytes, the gene expression of ki67 was increased in both cell types, the gene expression of tenomodulin was increased in both cell types at 100 ng/mL TIMP-1, and alkaline phosphatase expression ALP rose significantly in rbASCs. Electrospun TIMP-1/DP fibers had a ~5 μm diameter, a ~10 μm pore size, and a mesh thickness of ~200 μm. TIMP-1/DP meshes were more hydrophilic than pure DP meshes. TIMP-1 was released from the meshes with a sustained release of up to 7 days. CONCLUSIONS TIMP-1/DP tubes may be used to modulate the fibrotic tissue reaction when applied around conventionally sutured tendon ruptures.
Collapse
Affiliation(s)
| | | | | | - Johanna Buschmann
- Department of Plastic Surgery and Hand Surgery, University Hospital Zurich, Sternwartstrasse 14, 8091 Zurich, Switzerland; (J.R.); (R.K.N.); (P.G.)
| |
Collapse
|
6
|
Wang Q, Li Y, Yao L, Li H, Zhang L, Wang Y, Li J, Chen T, Chai K, Gao J, Gao J, Su L, Li X. High-affinity ssDNA aptamer and chemiluminescent aptasensor for TIMP-1 detection in human serum. ANAL SCI 2025; 41:119-126. [PMID: 39742443 PMCID: PMC11750923 DOI: 10.1007/s44211-024-00673-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2024] [Accepted: 09/17/2024] [Indexed: 01/03/2025]
Abstract
TIMP-1 (Tissue Inhibitor of Metalloproteinases-1) is a protein involved in regulating extracellular matrix (ECM) degradation. It is recognized as a significant biomarker for cancer diagnosis. This study aimed to develop and characterize a single-stranded DNA (ssDNA) aptamer targeting human TIMP-1 protein with high affinity and specificity. A magnetic beads-based SELEX process combined with qPCR was used to select aptamers over seven rounds. The enriched ssDNA library was analyzed using high-throughput sequencing to identify candidate sequences, and these sequences were characterized using surface plasmon resonance (SPR) and binding assays to evaluate their affinity and specificity. The selected ssDNA aptamer demonstrated a dissociation equilibrium constant (KD) of 0.41 nM and a very slow off-rate, enabling effective capture of TIMP-1 in serum samples. Furthermore, a chemiluminescent aptasensor was developed for TIMP-1 detection, which exhibited high specificity and a broad linear detection range from 1 to 500 ng/mL in human serum. The developed ssDNA aptamer targeting TIMP-1 shows high affinity and specificity, and the chemiluminescent aptasensor demonstrates promising potential for clinical diagnosis of TIMP-1 levels in human serum.
Collapse
Affiliation(s)
- Qin Wang
- The Third People's Hospital of Zhengzhou, Zhengzhou, 450000, China
| | - Yanli Li
- The First People's Hospital of Shangqiu, Shangqiu, 476000, China
| | - Lige Yao
- The Third People's Hospital of Zhengzhou, Zhengzhou, 450000, China
| | - Huiqin Li
- The Third People's Hospital of Zhengzhou, Zhengzhou, 450000, China
| | - Liuyan Zhang
- The Third People's Hospital of Zhengzhou, Zhengzhou, 450000, China
| | - Yingjie Wang
- The Third People's Hospital of Zhengzhou, Zhengzhou, 450000, China
| | - Jiayin Li
- The Third People's Hospital of Zhengzhou, Zhengzhou, 450000, China
| | - Tian Chen
- The Third People's Hospital of Zhengzhou, Zhengzhou, 450000, China
| | - Kun Chai
- Hangzhou Cosmos Wisdom Mass Spectrometry Center of Zhejiang University Medical School, Hangzhou, 311200, China
| | - Junli Gao
- Hangzhou Cosmos Wisdom Mass Spectrometry Center of Zhejiang University Medical School, Hangzhou, 311200, China
| | - Junshun Gao
- Hangzhou Cosmos Wisdom Mass Spectrometry Center of Zhejiang University Medical School, Hangzhou, 311200, China
| | - Li Su
- The Third People's Hospital of Zhengzhou, Zhengzhou, 450000, China.
| | - Xueming Li
- Hangzhou Cosmos Wisdom Mass Spectrometry Center of Zhejiang University Medical School, Hangzhou, 311200, China.
| |
Collapse
|
7
|
Mathias CB, Badaró RF, Bautz WG, da Gama-de-Souza LN. How malocclusion interferes with tissue inhibitor of metalloproteinase-1 expression and morphology of the articular cartilage of the mandible in female rats. Arch Oral Biol 2025; 169:106117. [PMID: 39471769 DOI: 10.1016/j.archoralbio.2024.106117] [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: 06/20/2024] [Revised: 10/08/2024] [Accepted: 10/19/2024] [Indexed: 11/01/2024]
Abstract
OBJECTIVE The purpose of this study was to investigate morphological alterations and tissue inhibitor of metalloproteinase-1 expression in the articular cartilage of the mandible under conditions of experimentally induced malocclusion. DESIGN Twenty-four 8-week-old female Wistar rats were used and divided into control and experimental groups with two different treatment periods (2 and 4 weeks). Sagittal malocclusions were orthodontically created, causing mesial movement of the first molars and distalization of the third molars unilaterally and on opposite sides of the arches. Sagittal sections of the articular cartilage of the mandible were subjected to hematoxylin and eosin and immunohistochemistry for tissue inhibitor of metalloproteinase-1. Chi-square and MannWhitney U tests were applied. RESULTS Animals treated for 2 and 4 weeks showed morphological alterations in articular cartilage of the mandible. The main findings were thickening of the posterior third, layer derangement, osteoclast activity and osteophyte formation. Among the cellular aspects, the presence of chondrocytes with condensed nuclei and cytoplasm reduction were observed. The enzyme in control animals was observed only in the mature layer. Treated animals showed immunopositive cells in the proliferative and mature layers, and in the 2-week treated group, the posterior third of the cartilage had more immunolabeled cells than control (P=0.0291). CONCLUSIONS The occlusal disorder caused morphological changes in articular cartilage of the mandible, probably due to the attempt to adapt to the new condition. Tissue inhibitor of metalloproteinase-1 expression may play a role as an initial modulator in the biological events observed in articular cartilage of the mandible.
Collapse
Affiliation(s)
- Carolina Brioschi Mathias
- Federal University of Espírito Santo, Marechal Campos Avenue, 1468, Maruípe, Vitória, ES ZIP CODE 29043-900, Brazil.
| | - Rebeca Ferreira Badaró
- Graduate Program in Dental Sciences, Federal University of Espírito Santo, Marechal Campos avenue, 1468, Maruípe, Vitória, ES ZIP CODE 29043-900, Brazil.
| | - Willian Grassi Bautz
- Department of Morphology, Federal University of Espírito Santo, Marechal Campos Avenue, 1468, Maruípe, Vitória, ES ZIP CODE 29043-900, Brazil.
| | - Leticia Nogueira da Gama-de-Souza
- Department of Morphology, Graduate Program in Dental Sciences, Federal University of Espírito Santo, Marechal Campos Avenue, 1468, Maruípe, Vitória, ES ZIP CODE 29043, Brazil.
| |
Collapse
|
8
|
Dutta S, Zhu Y, Almuntashiri S, Peh HY, Zuñiga J, Zhang D, Somanath PR, Ramírez G, Irineo-Moreno V, Jiménez-Juárez F, López-Salinas K, Regino N, Campero P, Crocker SJ, Owen CA, Wang X. PDGFRα-positive cell-derived TIMP-1 modulates adaptive immune responses to influenza A viral infection. Am J Physiol Lung Cell Mol Physiol 2025; 328:L60-L74. [PMID: 39585242 PMCID: PMC11905806 DOI: 10.1152/ajplung.00104.2024] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2024] [Revised: 10/16/2024] [Accepted: 11/05/2024] [Indexed: 11/26/2024] Open
Abstract
Tissue inhibitor of metalloproteinases-1 (TIMP-1) is a physiologic inhibitor of the matrix metalloproteinases (MMPs), but little is known about the role of TIMP-1 in regulating the pathogenesis of influenza A virus (IAV) infection. Here, we performed both in vivo and in vitro experiments to investigate the regulation and function of TIMP-1 during IAV infection. Specifically, plasma levels of TIMP-1 are significantly increased in human subjects and wild-type (WT) mice infected with 2009 H1N1 IAV compared with levels in uninfected controls. Also, TIMP-1 is strikingly upregulated in PDGFRα positive (PDGFRα+) cells in IAV-infected murine lungs as demonstrated using conditional KO (cKO) mice with a specific deletion of Timp-1 in PDGFRα+ cells. Our in vitro data indicated that TIMP-1 is induced by transforming growth factor-β (TGF-β) during lipofibroblasts (lipoFBs)-to-myofibroblast (myoFB) transdifferentiation. Timp-1 deficiency protects mice from H1N1 IAV-induced weight loss, mortality, and lung injury. IAV-infected Timp-1-deficient mice showed increased macrophages, and B and T cell counts in bronchoalveolar lavage (BAL) on day 7 postinfection (p.i.), but reduced BAL neutrophil counts. Increased Cxcl12 levels were detected in both BAL cells and lungs from Timp-1-deficient mice on day 3 p.i. Taken together, our data strongly link TIMP-1 to IAV pathogenesis. We identified that PDGFRα-lineage cells are the main cellular source of elevated TIMP-1 during IAV infection. Loss of Timp-1 attenuates IAV-induced mortality and promotes T and B cell recruitment. Thus, TIMP-1 may be a novel therapeutic target for IAV infection.NEW & NOTEWORTHY Our data strongly link tissue inhibitor of metalloproteinases-1 (TIMP-1) to influenza A virus (IAV) pathogenesis. TIMP-1 is highly increased in PDGFRα-lineage cells during IAV infection. Transforming growth factor-β (TGF-β) induces TIMP-1 during lipofibroblast (lipoFB)-to- myofibroblast (myoFB) transdifferentiation. Timp-1 deficiency protects mice from H1N1 IAV-induced weight loss, mortality, and lung injury. TIMP-1 may be a novel therapeutic target for IAV infection.
Collapse
Affiliation(s)
- Saugata Dutta
- Clinical and Experimental Therapeutics, College of Pharmacy, University of Georgia, Augusta, Georgia, United States
- Charlie Norwood VA Medical Center, Augusta, Georgia, United States
| | - Yin Zhu
- Clinical and Experimental Therapeutics, College of Pharmacy, University of Georgia, Augusta, Georgia, United States
- Charlie Norwood VA Medical Center, Augusta, Georgia, United States
| | - Sultan Almuntashiri
- Clinical and Experimental Therapeutics, College of Pharmacy, University of Georgia, Augusta, Georgia, United States
- Charlie Norwood VA Medical Center, Augusta, Georgia, United States
- Department of Clinical Pharmacy, College of Pharmacy, University of Hail, Hail, Saudi Arabia
| | - Hong Yong Peh
- Division of Pulmonary and Critical Care Medicine, Brigham and Women's Hospital, and Harvard Medical School, Boston, Massachusetts, United States
| | - Joaquin Zuñiga
- Laboratory of Immunobiology and Genetics and Intensive Care Unit, Instituto Nacional de Enfermedades Respiratorias Ismael Cosío Villegas, Mexico City, Mexico
- Tecnologico de Monterrey, School of Medicine and Health Sciences, Mexico City, Mexico
| | - Duo Zhang
- Clinical and Experimental Therapeutics, College of Pharmacy, University of Georgia, Augusta, Georgia, United States
- Charlie Norwood VA Medical Center, Augusta, Georgia, United States
- Tecnologico de Monterrey, School of Medicine and Health Sciences, Mexico City, Mexico
- Department of Medicine, Medical College of Georgia, Augusta University, Augusta, Georgia, United States
| | - Payaningal R Somanath
- Clinical and Experimental Therapeutics, College of Pharmacy, University of Georgia, Augusta, Georgia, United States
- Charlie Norwood VA Medical Center, Augusta, Georgia, United States
| | - Gustavo Ramírez
- Laboratory of Immunobiology and Genetics and Intensive Care Unit, Instituto Nacional de Enfermedades Respiratorias Ismael Cosío Villegas, Mexico City, Mexico
| | - Valeria Irineo-Moreno
- Laboratory of Immunobiology and Genetics and Intensive Care Unit, Instituto Nacional de Enfermedades Respiratorias Ismael Cosío Villegas, Mexico City, Mexico
- Tecnologico de Monterrey, School of Medicine and Health Sciences, Mexico City, Mexico
| | - Fabiola Jiménez-Juárez
- Laboratory of Immunobiology and Genetics and Intensive Care Unit, Instituto Nacional de Enfermedades Respiratorias Ismael Cosío Villegas, Mexico City, Mexico
- Tecnologico de Monterrey, School of Medicine and Health Sciences, Mexico City, Mexico
| | - Karen López-Salinas
- Laboratory of Immunobiology and Genetics and Intensive Care Unit, Instituto Nacional de Enfermedades Respiratorias Ismael Cosío Villegas, Mexico City, Mexico
- Tecnologico de Monterrey, School of Medicine and Health Sciences, Mexico City, Mexico
| | - Nora Regino
- Laboratory of Immunobiology and Genetics and Intensive Care Unit, Instituto Nacional de Enfermedades Respiratorias Ismael Cosío Villegas, Mexico City, Mexico
- Tecnologico de Monterrey, School of Medicine and Health Sciences, Mexico City, Mexico
| | - Paloma Campero
- Laboratory of Immunobiology and Genetics and Intensive Care Unit, Instituto Nacional de Enfermedades Respiratorias Ismael Cosío Villegas, Mexico City, Mexico
| | - Stephen J Crocker
- Department of Neuroscience, University of Connecticut School of Medicine, Farmington, Connecticut, United States
| | - Caroline A Owen
- Division of Pulmonary and Critical Care Medicine, Brigham and Women's Hospital, and Harvard Medical School, Boston, Massachusetts, United States
| | - Xiaoyun Wang
- Clinical and Experimental Therapeutics, College of Pharmacy, University of Georgia, Augusta, Georgia, United States
- Charlie Norwood VA Medical Center, Augusta, Georgia, United States
- Division of Pulmonary and Critical Care Medicine, Brigham and Women's Hospital, and Harvard Medical School, Boston, Massachusetts, United States
| |
Collapse
|
9
|
Peterson R, Nothnick WB. Assessing Gelatinase Activity in Normal and Disease Uterine Tissue and Cells Via Gelatin Zymography. Methods Mol Biol 2025; 2918:229-237. [PMID: 40261627 DOI: 10.1007/978-1-0716-4482-9_19] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/24/2025]
Abstract
Matrix metalloproteinases (MMPs) are critical for the maintenance and remodeling of the extracellular matrix (ECM) under normal physiological conditions such as pregnancy and wound healing. However, an increase of MMPs in uterine diseases, such as adenomyosis, endometrial cancer, endometriosis, and uterine fibroids, has been observed and suspected to contribute to important pathophysiology phenotypes like invasion and migration. Of note, MMP-2 (also referred to as gelatinase A) and MMP-9 (also referred to as gelatinase B) are common gelatinases that have demonstrated increased activity in uterine diseases and in several cancer types, such as breast cancer, to promote cancerous phenotypes like increased invasion and migration. In-gel zymography is a useful technique for the detection of MMP activity via degradation of gelatin in gelatin-based gels. Using zymography, it is possible to assess the activity levels of MMP-2 and MMP-9 via gelatin degradation during the zymography process. Here, we will describe the process of zymography and assessment of MMP activity levels (MMP-2 and MMP-9) for both uterine tissues and cancerous cell lines.
Collapse
Affiliation(s)
- Riley Peterson
- Departments of Cellular Biology and Physiology and Obstetrics and Gynecology, Center for Reproductive Sciences, Institute for Reproductive and Developmental Sciences, University of Kansas Medical Center, Kansas City, KS, USA
| | - Warren B Nothnick
- Departments of Cellular Biology and Physiology and Obstetrics and Gynecology, Center for Reproductive Sciences, Institute for Reproductive and Developmental Sciences, University of Kansas Medical Center, Kansas City, KS, USA.
| |
Collapse
|
10
|
Tang T, Chen H, Hu L, Ye J, Jing C, Xu C, Wu X, Chen Y, Chen Z, Zhou H, Fan L, Fu X, Qian C, Chen J, Tan Z, Liu J, Zeng H, Chen G, Liu F. TIMP1 protects against blood-brain barrier disruption after subarachnoid haemorrhage by inhibiting ubiquitination of astrocytic β1-integrin. Stroke Vasc Neurol 2024; 9:671-684. [PMID: 38485231 PMCID: PMC11791639 DOI: 10.1136/svn-2023-002956] [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: 10/28/2023] [Accepted: 02/07/2024] [Indexed: 01/02/2025] Open
Abstract
BACKGROUND Astrocytes regulate blood-brain barrier (BBB) integrity, whereas subarachnoid haemorrhage (SAH) results in astrocyte dysregulation and BBB disruption. Here, we explored the involvement of tissue inhibitor of matrix metalloprotease-1 (TIMP1) in astrocyte-mediated BBB protection during SAH, along with its underlying mechanisms. METHODS C57BL/6J mice were used to establish a model of SAH. The effects of TIMP1 on SAH outcomes were analysed by intraperitoneal injection of recombinant mouse TIMP1 protein (rm-TIMP1; 250 µg/kg). The roles of TIMP1 and its effector β1-integrin on astrocytes were observed by in vivo transduction with astrocyte-targeted adeno-associated virus carrying TIMP1 overexpression plasmid or β1-integrin RNAi. The molecular mechanisms underlying TIMP1 and β1-integrin interactions were explored in primary cultured astrocytes stimulated with red blood cells (RBCs). RESULTS TIMP1 was upregulated after SAH. Administration of rm-TIMP1 mitigated SAH-induced early brain injury (EBI) in male and female mice. TIMP1 was primarily expressed in astrocytes; its overexpression in astrocytes led to increased β1-integrin expression in astrocytes, along with the preservation of astrocytic endfoot attachment to the endothelium and subsequent recovery of endothelial tight junctions. All of these effects were reversed by the knockdown of β1-integrin in astrocytes. Molecular analysis showed that TIMP1 overexpression decreased the RBC-induced ubiquitination of β1-integrin; this effect was partially achieved by inhibiting the interaction between β1-integrin and the E3 ubiquitin ligase Trim21. CONCLUSION TIMP1 inhibits the interaction between β1-integrin and Trim21 in astrocytes, thereby rescuing the ubiquitination of astrocytic β1-integrin. It subsequently restores interactions between astrocytic endfeet and the endothelium, as well as BBB integrity, eventually mitigating SAH-induced EBI.
Collapse
Affiliation(s)
- Tianchi Tang
- Neurosurgery, Zhejiang University School of Medicine Second Affiliated Hospital, Hangzhou, Zhejiang, China
- Key Laboratory of Precise Treatment and Clinical Translational Research of Neurological Diseases, Hangzhou, China
| | - Huaijun Chen
- Neurosurgery, Zhejiang University School of Medicine Second Affiliated Hospital, Hangzhou, Zhejiang, China
- Key Laboratory of Precise Treatment and Clinical Translational Research of Neurological Diseases, Hangzhou, China
| | - Libin Hu
- Neurosurgery, Zhejiang University School of Medicine Second Affiliated Hospital, Hangzhou, Zhejiang, China
- Key Laboratory of Precise Treatment and Clinical Translational Research of Neurological Diseases, Hangzhou, China
| | - Jingya Ye
- Zhejiang University School of Medicine, Hangzhou, Zhejiang, China
| | - Chaohui Jing
- Shanghai Jiaotong University School of Medicine Xinhua Hospital, Shanghai, China
| | - Chaoran Xu
- Neurosurgery, Zhejiang University School of Medicine Second Affiliated Hospital, Hangzhou, Zhejiang, China
- Key Laboratory of Precise Treatment and Clinical Translational Research of Neurological Diseases, Hangzhou, China
| | - Xinyan Wu
- Neurosurgery, Zhejiang University School of Medicine Second Affiliated Hospital, Hangzhou, Zhejiang, China
- Key Laboratory of Precise Treatment and Clinical Translational Research of Neurological Diseases, Hangzhou, China
| | - Yike Chen
- Neurosurgery, Zhejiang University School of Medicine Second Affiliated Hospital, Hangzhou, Zhejiang, China
- Key Laboratory of Precise Treatment and Clinical Translational Research of Neurological Diseases, Hangzhou, China
| | - Zihang Chen
- Neurosurgery, Zhejiang University School of Medicine Second Affiliated Hospital, Hangzhou, Zhejiang, China
- Key Laboratory of Precise Treatment and Clinical Translational Research of Neurological Diseases, Hangzhou, China
| | - Hang Zhou
- Neurosurgery, Zhejiang University School of Medicine Second Affiliated Hospital, Hangzhou, Zhejiang, China
- Key Laboratory of Precise Treatment and Clinical Translational Research of Neurological Diseases, Hangzhou, China
| | - Linfeng Fan
- Neurosurgery, Zhejiang University School of Medicine Second Affiliated Hospital, Hangzhou, Zhejiang, China
- Key Laboratory of Precise Treatment and Clinical Translational Research of Neurological Diseases, Hangzhou, China
| | - Xiongjie Fu
- Neurosurgery, Zhejiang University School of Medicine Second Affiliated Hospital, Hangzhou, Zhejiang, China
- Key Laboratory of Precise Treatment and Clinical Translational Research of Neurological Diseases, Hangzhou, China
| | - Cong Qian
- Neurosurgery, Zhejiang University School of Medicine Second Affiliated Hospital, Hangzhou, Zhejiang, China
| | - Jingsen Chen
- Neurosurgery, Zhejiang University School of Medicine Second Affiliated Hospital, Hangzhou, Zhejiang, China
| | - Zhongju Tan
- Zhejiang University School of Medicine First Affiliated Hospital, Hangzhou, Zhejiang, China
| | - Jing Liu
- Department of Nursing, Zhejiang University School of Medicine Second Affiliated Hospital, Hangzhou, Zhejiang, China
| | - Hanhai Zeng
- Neurosurgery, Zhejiang University School of Medicine Second Affiliated Hospital, Hangzhou, Zhejiang, China
- Key Laboratory of Precise Treatment and Clinical Translational Research of Neurological Diseases, Hangzhou, China
| | - Gao Chen
- Neurosurgery, Zhejiang University School of Medicine Second Affiliated Hospital, Hangzhou, Zhejiang, China
- Key Laboratory of Precise Treatment and Clinical Translational Research of Neurological Diseases, Hangzhou, China
| | - Fuyi Liu
- Neurosurgery, Zhejiang University School of Medicine Second Affiliated Hospital, Hangzhou, Zhejiang, China
- Key Laboratory of Precise Treatment and Clinical Translational Research of Neurological Diseases, Hangzhou, China
| |
Collapse
|
11
|
Kimura T, Kruhlak M, Zhao L, Hwang E, Fozzatti L, Cheng SY. Combinatory actions of cytokines induce M2-like macrophages in anaplastic thyroid cancer. Am J Cancer Res 2024; 14:5812-5825. [PMID: 39803637 PMCID: PMC11711523 DOI: 10.62347/quwq3794] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2024] [Accepted: 12/15/2024] [Indexed: 01/16/2025] Open
Abstract
Anaplastic thyroid cancer (ATC) is a lethal endocrine malignancy. It has been shown that tumor-associated macrophages (TAMs) contribute to the aggressiveness of ATC. However, stimulatory factors that could facilitate the induction and infiltration of TAMs in the ATC tumor microenvironment (TME) are not fully elucidated. In this study, we used a human leukemia monocytic cell line (THP-1) to study the differentiation of THP-1 into M2-like macrophages (M2) by conditioned media (CM) derived from each of the three human ATC cells: 8505C, THJ-11T (11T), and THJ-16T (16T). The capacity of CM to induce M2 was in the order of 16T>8505C>11T cells as determined by the expression of M2 markers (CD163, CD204, and CCL13). Cytokine arrays and ELISA assays revealed five commonly enriched cytokines (IL-6, IL-8, MCP-1, TIMP-1, and TGF-β1) in the CM derived from each of the three ATC cells. These cytokines, individually, had weak activity, but together, they mimicked full CM activity in the induction of M2. Further, they collaboratively activated STAT3, ERK, and PI3K-AKT signaling to facilitate the induction of M2 as found in CM. Importantly, we found that the CM-induced M2 could secrete soluble growth factors to promote ATC cell proliferation as evidenced by the increased Ki-67, cMYC, and cyclin D1 protein levels. Our studies identified the major stimulatory cytokines which acted collaboratively to induce M2 in the TME. Importantly, the present studies indicate that when using inhibitors to target TAMs, combination therapies would be required for effective treatment of ATC.
Collapse
Affiliation(s)
- Takahito Kimura
- Laboratory of Molecular Biology, National Cancer Institute, National Institutes of HealthBethesda, MD 20892, USA
| | - Michael Kruhlak
- Laboratory of Cancer Biology and Genetics, National Cancer Institute, National Institutes of HealthBethesda, MD 20892, USA
| | - Li Zhao
- Laboratory of Molecular Biology, National Cancer Institute, National Institutes of HealthBethesda, MD 20892, USA
| | - Eunmi Hwang
- Laboratory of Molecular Biology, National Cancer Institute, National Institutes of HealthBethesda, MD 20892, USA
| | - Laura Fozzatti
- Departamento de Bioquímica Clínica, Facultad de Ciencias Químicas, Universidad Nacional de CórdobaCórdoba 5000, Argentina
| | - Sheue-Yann Cheng
- Laboratory of Molecular Biology, National Cancer Institute, National Institutes of HealthBethesda, MD 20892, USA
| |
Collapse
|
12
|
Jin Y, Yang M, Zhao W, Liu M, Fang W, Wang Y, Gao G, Wang Y, Fu Q. Scaffold-based tissue engineering strategies for urethral repair and reconstruction. Biofabrication 2024; 17:012003. [PMID: 39433068 DOI: 10.1088/1758-5090/ad8965] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2024] [Accepted: 10/21/2024] [Indexed: 10/23/2024]
Abstract
Urethral strictures are common in urology; however, the reconstruction of long urethral strictures remains challenging. There are still unavoidable limitations in the clinical application of grafts for urethral injuries, which has facilitated the advancement of urethral tissue engineering. Tissue-engineered urethral scaffolds that combine cells or bioactive factors with a biomaterial to mimic the native microenvironment of the urethra, offer a promising approach to urethral reconstruction. Despite the recent rapid development of tissue engineering materials and techniques, a consensus on the optimal strategy for urethral repair and reconstruction is still lacking. This review aims to collect the achievements of urethral tissue engineering in recent years and to categorize and summarize them to shed new light on their design. Finally, we visualize several important future directions for urethral repair and reconstruction.
Collapse
Affiliation(s)
- Yangwang Jin
- Department of Urology, Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai Eastern Institute of Urologic Reconstruction, Shanghai Jiao Tong University, Shanghai 200233, People's Republic of China
| | - Ming Yang
- Department of Urology, Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai Eastern Institute of Urologic Reconstruction, Shanghai Jiao Tong University, Shanghai 200233, People's Republic of China
| | - Weixin Zhao
- Wake Forest Institute for Regenerative Medicine, Winston Salem, NC, United States of America
| | - Meng Liu
- Department of Urology, Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai Eastern Institute of Urologic Reconstruction, Shanghai Jiao Tong University, Shanghai 200233, People's Republic of China
| | - Wenzhuo Fang
- Department of Urology, Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai Eastern Institute of Urologic Reconstruction, Shanghai Jiao Tong University, Shanghai 200233, People's Republic of China
| | - Yuhui Wang
- Department of Urology, Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai Eastern Institute of Urologic Reconstruction, Shanghai Jiao Tong University, Shanghai 200233, People's Republic of China
| | - Guo Gao
- Key Laboratory for Thin Film and Micro Fabrication of the Ministry of Education, School of Sensing Science and Engineering, School of Electronic Information and Electrical Engineering, Shanghai Jiao Tong University, Shanghai 200240, People's Republic of China
| | - Ying Wang
- Department of Urology, Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai Eastern Institute of Urologic Reconstruction, Shanghai Jiao Tong University, Shanghai 200233, People's Republic of China
| | - Qiang Fu
- Department of Urology, Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai Eastern Institute of Urologic Reconstruction, Shanghai Jiao Tong University, Shanghai 200233, People's Republic of China
| |
Collapse
|
13
|
Mishra RR, Nielsen BE, Trudrung MA, Lee S, Bolstad LJ, Hellenbrand DJ, Hanna AS. The Effect of Tissue Inhibitor of Metalloproteinases on Scar Formation after Spinal Cord Injury. Cells 2024; 13:1547. [PMID: 39329731 PMCID: PMC11430430 DOI: 10.3390/cells13181547] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2024] [Revised: 09/09/2024] [Accepted: 09/10/2024] [Indexed: 09/28/2024] Open
Abstract
Spinal cord injury (SCI) often results in permanent loss of motor and sensory function. After SCI, the blood-spinal cord barrier (BSCB) is disrupted, causing the infiltration of neutrophils and macrophages, which secrete several kinds of cytokines, as well as matrix metalloproteinases (MMPs). MMPs are proteases capable of degrading various extracellular matrix (ECM) proteins, as well as many non-matrix substrates. The tissue inhibitor of MMPs (TIMP)-1 is significantly upregulated post-SCI and operates via MMP-dependent and MMP-independent pathways. Through the MMP-dependent pathway, TIMP-1 directly reduces inflammation and destruction of the ECM by binding and blocking the catalytic domains of MMPs. Thus, TIMP-1 helps preserve the BSCB and reduces immune cell infiltration. The MMP-independent pathway involves TIMP-1's cytokine-like functions, in which it binds specific TIMP surface receptors. Through receptor binding, TIMP-1 can stimulate the proliferation of several types of cells, including keratinocytes, aortic smooth muscle cells, skin epithelial cells, corneal epithelial cells, and astrocytes. TIMP-1 induces astrocyte proliferation, modulates microglia activation, and increases myelination and neurite extension in the central nervous system (CNS). In addition, TIMP-1 also regulates apoptosis and promotes cell survival through direct signaling. This review provides a comprehensive assessment of TIMP-1, specifically regarding its contribution to inflammation, ECM remodeling, and scar formation after SCI.
Collapse
Affiliation(s)
- Raveena R. Mishra
- Department of Neurosurgery, School of Medicine and Public Health, University of Wisconsin-Madison, Madison, WI 53706, USA; (R.R.M.); (B.E.N.); (M.A.T.); (S.L.); (L.J.B.)
| | - Brooke E. Nielsen
- Department of Neurosurgery, School of Medicine and Public Health, University of Wisconsin-Madison, Madison, WI 53706, USA; (R.R.M.); (B.E.N.); (M.A.T.); (S.L.); (L.J.B.)
| | - Melissa A. Trudrung
- Department of Neurosurgery, School of Medicine and Public Health, University of Wisconsin-Madison, Madison, WI 53706, USA; (R.R.M.); (B.E.N.); (M.A.T.); (S.L.); (L.J.B.)
| | - Samuel Lee
- Department of Neurosurgery, School of Medicine and Public Health, University of Wisconsin-Madison, Madison, WI 53706, USA; (R.R.M.); (B.E.N.); (M.A.T.); (S.L.); (L.J.B.)
| | - Luke J. Bolstad
- Department of Neurosurgery, School of Medicine and Public Health, University of Wisconsin-Madison, Madison, WI 53706, USA; (R.R.M.); (B.E.N.); (M.A.T.); (S.L.); (L.J.B.)
| | - Daniel J. Hellenbrand
- Department of Neurosurgery, School of Medicine and Public Health, University of Wisconsin-Madison, Madison, WI 53706, USA; (R.R.M.); (B.E.N.); (M.A.T.); (S.L.); (L.J.B.)
- Department of Biomedical Engineering, University of Wisconsin-Madison, Madison, WI 53706, USA
| | - Amgad S. Hanna
- Department of Neurosurgery, School of Medicine and Public Health, University of Wisconsin-Madison, Madison, WI 53706, USA; (R.R.M.); (B.E.N.); (M.A.T.); (S.L.); (L.J.B.)
- Department of Biomedical Engineering, University of Wisconsin-Madison, Madison, WI 53706, USA
| |
Collapse
|
14
|
Jung S, Cheong S, Lee Y, Lee J, Lee J, Kwon MS, Oh YS, Kim T, Ha S, Kim SJ, Jo DH, Ko J, Jeon NL. Integrating Vascular Phenotypic and Proteomic Analysis in an Open Microfluidic Platform. ACS NANO 2024; 18:24909-24928. [PMID: 39208278 PMCID: PMC11394367 DOI: 10.1021/acsnano.4c05537] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/26/2024] [Revised: 08/19/2024] [Accepted: 08/20/2024] [Indexed: 09/04/2024]
Abstract
This research introduces a vascular phenotypic and proteomic analysis (VPT) platform designed to perform high-throughput experiments on vascular development. The VPT platform utilizes an open-channel configuration that facilitates angiogenesis by precise alignment of endothelial cells, allowing for a 3D morphological examination and protein analysis. We study the effects of antiangiogenic agents─bevacizumab, ramucirumab, cabozantinib, regorafenib, wortmannin, chloroquine, and paclitaxel─on cytoskeletal integrity and angiogenic sprouting, observing an approximately 50% reduction in sprouting at higher drug concentrations. Precise LC-MS/MS analyses reveal global protein expression changes in response to four of these drugs, providing insights into the signaling pathways related to the cell cycle, cytoskeleton, cellular senescence, and angiogenesis. Our findings emphasize the intricate relationship between cytoskeletal alterations and angiogenic responses, underlining the significance of integrating morphological and proteomic data for a comprehensive understanding of angiogenesis. The VPT platform not only advances our understanding of drug impacts on vascular biology but also offers a versatile tool for analyzing proteome and morphological features across various models beyond blood vessels.
Collapse
Affiliation(s)
- Sangmin Jung
- Department
of Mechanical Engineering, Seoul National
University, Seoul 08826, Republic
of Korea
| | - Sunghun Cheong
- Interdisciplinary
Program in Bioengineering, Seoul National
University, Seoul 08826, Republic
of Korea
| | - Yoonho Lee
- Interdisciplinary
Program in Bioengineering, Seoul National
University, Seoul 08826, Republic
of Korea
| | - Jungseub Lee
- Department
of Mechanical Engineering, Seoul National
University, Seoul 08826, Republic
of Korea
| | - Jihye Lee
- Target
Link Therapeutics, Inc., Seoul 04545, Republic
of Korea
| | - Min-Seok Kwon
- Target
Link Therapeutics, Inc., Seoul 04545, Republic
of Korea
- Department
of Public Health Science, Graduate School of Public Health, Seoul National University, Seoul 08826, Republic of Korea
| | - Young Sun Oh
- Department
of Mechanical Engineering, Seoul National
University, Seoul 08826, Republic
of Korea
- Target
Link Therapeutics, Inc., Seoul 04545, Republic
of Korea
| | - Taewan Kim
- Department
of Electrical and Computer Engineering, Seoul National University, Seoul 08826, Republic of Korea
| | - Sungjae Ha
- ProvaLabs,
Inc., Seoul 08826, Republic of Korea
| | - Sung Jae Kim
- Department
of Electrical and Computer Engineering, Seoul National University, Seoul 08826, Republic of Korea
- SOFT
Foundry, Seoul National University, Seoul 08826, Republic of Korea
- Inter-university
Semiconductor Research Center, Seoul National
University, Seoul 08826, Republic
of Korea
| | - Dong Hyun Jo
- Department
of Anatomy and Cell Biology, Seoul National
University College of Medicine, Seoul 03080, Republic of Korea
| | - Jihoon Ko
- Department
of BioNano Technology, Gachon University, Seongnam-si, Gyeonggi-do 13120, Republic
of Korea
| | - Noo Li Jeon
- Department
of Mechanical Engineering, Seoul National
University, Seoul 08826, Republic
of Korea
- Interdisciplinary
Program in Bioengineering, Seoul National
University, Seoul 08826, Republic
of Korea
- Institute
of Advanced Machines and Design, Seoul National
University, Seoul 08826, Republic
of Korea
- Qureator, Inc., San
Diego, California 92121, United States
| |
Collapse
|
15
|
Raftery RM, Gonzalez Vazquez AG, Walsh DP, Chen G, Laiva AL, Keogh MB, O'Brien FJ. Mobilizing Endogenous Progenitor Cells Using pSDF1α-Activated Scaffolds Accelerates Angiogenesis and Bone Repair in Critical-Sized Bone Defects. Adv Healthc Mater 2024; 13:e2401031. [PMID: 38850118 DOI: 10.1002/adhm.202401031] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2024] [Revised: 06/05/2024] [Indexed: 06/09/2024]
Abstract
Mobilizing endogenous progenitor cells to repair damaged tissue in situ has the potential to revolutionize the field of regenerative medicine, while the early establishment of a vascular network will ensure survival of newly generated tissue. In this study, a gene-activated scaffold containing a stromal derived factor 1α plasmid (pSDF1α), a pro-angiogenic gene that is also thought to be involved in the recruitment of mesenchymal stromal cells (MSCs) to sites of injury is described. It is shown that over-expression of SDF1α protein enhanced MSC recruitment and induced vessel-like structure formation by endothelial cells in vitro. When implanted subcutaneously, transcriptomic analysis reveals that endogenous MSCs are recruited and significant angiogenesis is stimulated. Just 1-week after implantation into a calvarial critical-sized bone defect, pSDF1α-activated scaffolds are recruited MSCs and rapidly activate angiogenic and osteogenic programs, upregulating Runx2, Dlx5, and Sp7. At the same time-point, pVEGF-activated scaffolds are recruited a variety of cell types, activating endochondral ossification. The early response induced by both scaffolds leads to complete bridging of the critical-sized bone defects within 4-weeks. The versatile cell-free gene-activated scaffold described in this study is capable of harnessing and enhancing the body's own regenerative capacity and has immense potential in a myriad of applications.
Collapse
Affiliation(s)
- Rosanne M Raftery
- Tissue Engineering Research Group, Department of Anatomy and Regenerative Medicine, Royal College of Surgeons in Ireland, Dublin, D02 YN77, Ireland
- Trinity Centre for Biomedical Engineering (TCBE), Trinity College Dublin, Dublin 2, Dublin, D02 PN40, Ireland
- Advanced Materials and Bioengineering Research Centre (AMBER), RCSI and TCD, Dublin, D02 YN77, Ireland
- iEd Hub and Department of Anatomy and Neuroscience, College of Medicine and Health, University College Cork, Cork, T12 CY82, Ireland
| | - Arlyng G Gonzalez Vazquez
- Tissue Engineering Research Group, Department of Anatomy and Regenerative Medicine, Royal College of Surgeons in Ireland, Dublin, D02 YN77, Ireland
- Trinity Centre for Biomedical Engineering (TCBE), Trinity College Dublin, Dublin 2, Dublin, D02 PN40, Ireland
- Advanced Materials and Bioengineering Research Centre (AMBER), RCSI and TCD, Dublin, D02 YN77, Ireland
| | - David P Walsh
- Tissue Engineering Research Group, Department of Anatomy and Regenerative Medicine, Royal College of Surgeons in Ireland, Dublin, D02 YN77, Ireland
- Trinity Centre for Biomedical Engineering (TCBE), Trinity College Dublin, Dublin 2, Dublin, D02 PN40, Ireland
- Advanced Materials and Bioengineering Research Centre (AMBER), RCSI and TCD, Dublin, D02 YN77, Ireland
- Translational Research in Nanomedical Devices, School of Pharmacy, Royal College of Surgeons in Ireland, Dublin, D02 YN77, Ireland
| | - Gang Chen
- Department of Physiology and Medical Physics, Centre for the Study of Neurological Disorders, Microsurgical Research and Training Facility (MRTF), Royal College of Surgeons in Ireland, Dublin, D02 YN77, Ireland
| | - Ashang L Laiva
- Tissue Engineering Research Group, Department of Anatomy and Regenerative Medicine, Royal College of Surgeons in Ireland, Dublin, D02 YN77, Ireland
- Tisse Engineering Research Group, Royal College of Surgeons in Ireland - Medical University of Bahrain, Adliya, Bahrain
| | - Michael B Keogh
- Tissue Engineering Research Group, Department of Anatomy and Regenerative Medicine, Royal College of Surgeons in Ireland, Dublin, D02 YN77, Ireland
- Tisse Engineering Research Group, Royal College of Surgeons in Ireland - Medical University of Bahrain, Adliya, Bahrain
| | - Fergal J O'Brien
- Tissue Engineering Research Group, Department of Anatomy and Regenerative Medicine, Royal College of Surgeons in Ireland, Dublin, D02 YN77, Ireland
- Trinity Centre for Biomedical Engineering (TCBE), Trinity College Dublin, Dublin 2, Dublin, D02 PN40, Ireland
- Advanced Materials and Bioengineering Research Centre (AMBER), RCSI and TCD, Dublin, D02 YN77, Ireland
| |
Collapse
|
16
|
Lin HC, Hsieh MH, Lo YL, Huang HY, Huang SW, Huang CD, Chang PJ, Lo CY, Lin TY, Fang YF, Lin SM, Lin CY, Tsai YH. IL-6 and TIMP-1 Correlated to Airway Pathogen Colonization and Predict Disease Severity in Patients with Non-Cystic Fibrosis Bronchiectasis. J Inflamm Res 2024; 17:5701-5709. [PMID: 39219819 PMCID: PMC11366244 DOI: 10.2147/jir.s465413] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2024] [Accepted: 08/19/2024] [Indexed: 09/04/2024] Open
Abstract
Background Non-cystic fibrosis bronchiectasis is associated with airway pathogen colonization. We planned to investigate the inflammatory markers in patients with different airway pathogens and their correlation with disease severity. Methods We enrolled patients aged between 20 and 75 from October 2021 to August 2022. All patients had sputum evaluation for bacterial and fungal cultures before enrollment, and were classified into four groups according to the culture results. Results Forty-four patients with non-CF bronchiectasis and six controls were enrolled and categorized as follows: Group 1, no pathogens identified in sputum cultures (n = 14); Group 2, positive fungal culture results (n = 18); Group 3, positive P. aeruginosa culture results (n = 7); and Group 4, positive culture results for both fungi and P. aeruginosa (n = 5). Group 4 had significantly higher serum defensin α1, IL-6 and tissue inhibitors of MMP (TIMP)-1 levels than group 1 patients. The serum levels of IL-6 and TIMP-1 were positively correlated with the FACED score and negatively correlated with distance-saturation product. Conclusion Significantly higher levels of serum IL-6 and TIMP-1 were found in the patients who had concomitant fungal and P. aeruginosa colonization, and were closely related to clinical severity and may have important roles in disease monitoring.
Collapse
Affiliation(s)
- Horng-Chyuan Lin
- Department of Pulmonary and Critical Care Medicine, Chang Gung Memorial Hospital at Linkou, Taoyuan, Taiwan
- College of Medicine, Chang Gung University, Taoyuan, Taiwan
- Department of Respiratory Therapy, Chang Gung Memorial Hospital at Linkou, Taoyuan, Taiwan
| | - Meng-heng Hsieh
- Department of Pulmonary and Critical Care Medicine, Chang Gung Memorial Hospital at Linkou, Taoyuan, Taiwan
- College of Medicine, Chang Gung University, Taoyuan, Taiwan
| | - Yu-Lun Lo
- Department of Pulmonary and Critical Care Medicine, Chang Gung Memorial Hospital at Linkou, Taoyuan, Taiwan
- College of Medicine, Chang Gung University, Taoyuan, Taiwan
| | - Hung-Yu Huang
- Department of Pulmonary and Critical Care Medicine, Chang Gung Memorial Hospital at Linkou, Taoyuan, Taiwan
- College of Medicine, Chang Gung University, Taoyuan, Taiwan
| | - Shih-Wei Huang
- Department of Pulmonary and Critical Care Medicine, Chang Gung Memorial Hospital at Linkou, Taoyuan, Taiwan
- College of Medicine, Chang Gung University, Taoyuan, Taiwan
| | - Chien-Da Huang
- Department of Pulmonary and Critical Care Medicine, Chang Gung Memorial Hospital at Linkou, Taoyuan, Taiwan
- College of Medicine, Chang Gung University, Taoyuan, Taiwan
| | - Po-Jui Chang
- Department of Pulmonary and Critical Care Medicine, Chang Gung Memorial Hospital at Linkou, Taoyuan, Taiwan
- College of Medicine, Chang Gung University, Taoyuan, Taiwan
| | - Chun-Yu Lo
- Department of Pulmonary and Critical Care Medicine, Chang Gung Memorial Hospital at Linkou, Taoyuan, Taiwan
- College of Medicine, Chang Gung University, Taoyuan, Taiwan
| | - Ting-Yu Lin
- Department of Pulmonary and Critical Care Medicine, Chang Gung Memorial Hospital at Linkou, Taoyuan, Taiwan
- College of Medicine, Chang Gung University, Taoyuan, Taiwan
| | - Yueh-Fu Fang
- Department of Pulmonary and Critical Care Medicine, Chang Gung Memorial Hospital at Linkou, Taoyuan, Taiwan
- College of Medicine, Chang Gung University, Taoyuan, Taiwan
| | - Shu-Min Lin
- Department of Pulmonary and Critical Care Medicine, Chang Gung Memorial Hospital at Linkou, Taoyuan, Taiwan
- College of Medicine, Chang Gung University, Taoyuan, Taiwan
| | - Chun-Yu Lin
- Department of Pulmonary and Critical Care Medicine, Chang Gung Memorial Hospital at Linkou, Taoyuan, Taiwan
- College of Medicine, Chang Gung University, Taoyuan, Taiwan
| | - Ying-Huang Tsai
- Department of Pulmonary and Critical Care Medicine, Chang Gung Memorial Hospital at Linkou, Taoyuan, Taiwan
- College of Medicine, Chang Gung University, Taoyuan, Taiwan
- Department of Pulmonary and Critical Care Medicine, Xiamen Chang Gung Hospital, Xiamen, 361028, People’s Republic of China
| |
Collapse
|
17
|
Yao Y, Tian G, Zhang J, Zhang S, Liu X, Hou J. Integrating bulk and single-cell sequencing reveals metastasis-associated CAFs in head and neck squamous cell carcinoma. Life Sci 2024; 351:122768. [PMID: 38851417 DOI: 10.1016/j.lfs.2024.122768] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2024] [Revised: 05/18/2024] [Accepted: 06/02/2024] [Indexed: 06/10/2024]
Abstract
AIMS Cancer-associated fibroblasts (CAFs) have been shown to promote the metastasis of head and neck squamous cell carcinoma (HNSCC), but the underlying mechanisms remain unclear. The purpose of this study is to identify gene in CAFs that are associated with metastasis and to preliminarily validate its impact on the metastasis of HNSCC. MATERIALS AND METHODS Scissor analysis was utilized to process single-cell and bulk RNA sequencing datasets, identifying genes associated with the metastasis of HNSCC through differential gene expression analysis. A risk model was constructed using LASSO regression analysis. Quantitative real time-PCR and Western blot were employed to measure mRNA and protein expressions, respectively. Multiplex immunohistochemistry (mIHC) was used to assess protein expression in CAFs. siRNA was utilized to achieve gene knockdown. CCK-8 and Transwell assays were employed to evaluate the biological characteristics of HNSCC cells. KEY FINDINGS Compare to the nonmetastatic primary CAFs (nmCAFs), tissue inhibitors of metalloproteinase-1 (TIMP1) was founded to be overexpressed in the cells and tissues of metastatic primary CAFs (mCAFs). Knocking down TIMP1 in CAFs can signifucantly inhibit the proliferation, invasion, and migration of HNSCC cells. SIGNIFICANCE CAFs facilitate HNSCC cell metastasis by upregulating TIMP1, highlighting TIMP1 as a potential therapeutic target in HNSCC metastasis management.
Collapse
Affiliation(s)
- Yihuan Yao
- Department of Oral and Maxillofacial Surgery, Hospital of Stomatology, Guanghua School of Stomatology, Sun Yat-sen University, 56 Ling-yuan west Street, Guangzhou 510000, China; Guangdong Provincial Key Laboratory of Stomatology, Sun Yat-sen University, Zhong Shan Er Road 74, Guangzhou 510080, China
| | - Guoli Tian
- Department of Oral and Maxillofacial Surgery, Hospital of Stomatology, Guanghua School of Stomatology, Sun Yat-sen University, 56 Ling-yuan west Street, Guangzhou 510000, China; Guangdong Provincial Key Laboratory of Stomatology, Sun Yat-sen University, Zhong Shan Er Road 74, Guangzhou 510080, China
| | - Jiaqiang Zhang
- Department of Oral and Maxillofacial Surgery, Hospital of Stomatology, Guanghua School of Stomatology, Sun Yat-sen University, 56 Ling-yuan west Street, Guangzhou 510000, China; Guangdong Provincial Key Laboratory of Stomatology, Sun Yat-sen University, Zhong Shan Er Road 74, Guangzhou 510080, China
| | - Shuaiyuan Zhang
- Department of Oral and Maxillofacial Surgery, Hospital of Stomatology, Guanghua School of Stomatology, Sun Yat-sen University, 56 Ling-yuan west Street, Guangzhou 510000, China; Guangdong Provincial Key Laboratory of Stomatology, Sun Yat-sen University, Zhong Shan Er Road 74, Guangzhou 510080, China
| | - Xiaoyong Liu
- Department of Oral and Maxillofacial Surgery, Hospital of Stomatology, Guanghua School of Stomatology, Sun Yat-sen University, 56 Ling-yuan west Street, Guangzhou 510000, China; Guangdong Provincial Key Laboratory of Stomatology, Sun Yat-sen University, Zhong Shan Er Road 74, Guangzhou 510080, China
| | - Jingsong Hou
- Department of Oral and Maxillofacial Surgery, Hospital of Stomatology, Guanghua School of Stomatology, Sun Yat-sen University, 56 Ling-yuan west Street, Guangzhou 510000, China; Guangdong Provincial Key Laboratory of Stomatology, Sun Yat-sen University, Zhong Shan Er Road 74, Guangzhou 510080, China.
| |
Collapse
|
18
|
Knapp S, Bolko L, Servettaz A, Didier K. [Eosinophilic fasciitis: From pathophysiology to therapeutics]. Rev Med Interne 2024; 45:488-497. [PMID: 38519306 DOI: 10.1016/j.revmed.2024.03.006] [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: 11/23/2023] [Revised: 02/27/2024] [Accepted: 03/07/2024] [Indexed: 03/24/2024]
Abstract
Eosinophilic fasciitis (EF) is a rare connective tissue disorder characterized by painful edema and induration of the limbs and trunk, likely associated with hypereosinophilia and hypergammaglobulinemia. EF causes arthralgia and range of motion limitation, leading to significant functional impairment and poor quality of life. Since its description by Shulman in 1974, over 300 cases have been reported. We present here a review of the latest diagnostic, pathophysiological and therapeutic developments in this disease. Magnetic resonance imaging appears useful to guide diagnosis and biopsy. Diagnosis is based on a deep skin biopsy involving the fascia, which will reveal edema, sclerofibrosis of the muscular fascia and subcutaneous tissue, and an inflammatory infiltrate sometimes composed of eosinophilic polynuclear cells. EF may occur in patients treated with immune checkpoint inhibitors and the diagnosis should be raised in case of cutaneous sclerosis in these patients. The pathophysiology of the disease remains poorly understood, and its management lacks randomized, controlled, blinded trials. First-line treatment consists in oral corticosteroid therapy, sometimes combined with an immunosuppressant, mainly methotrexate. A better understanding of the pathophysiology has opened new therapeutic perspectives and clarified the role of targeted therapies in the management of EF, such as interleukin-6 inhibitors, whose efficacy has been reported in several cases.
Collapse
Affiliation(s)
- S Knapp
- Service de médecine interne, maladie infectieuse et immunologie clinique, hôpital Robert-Debré, CHU de Reims, rue du Général-Kœnig, 51092 Reims cedex, France
| | - L Bolko
- Service de rhumatologie, hôpital Maison Blanche, CHU de Reims, Reims, France
| | - A Servettaz
- Service de médecine interne, maladie infectieuse et immunologie clinique, hôpital Robert-Debré, CHU de Reims, rue du Général-Kœnig, 51092 Reims cedex, France; EA 7509 IRMAIC, université de Reims Champagne-Ardenne, Reims, France
| | - K Didier
- Service de médecine interne, maladie infectieuse et immunologie clinique, hôpital Robert-Debré, CHU de Reims, rue du Général-Kœnig, 51092 Reims cedex, France; EA 7509 IRMAIC, université de Reims Champagne-Ardenne, Reims, France.
| |
Collapse
|
19
|
Riazi G, Brizais C, Garali I, Al-rifai R, Quelquejay H, Monceau V, Vares G, Ould-Boukhitine L, Aubeleau D, Gilain F, Gloaguen C, Dos Santos M, Ait-Oufella H, Ebrahimian T. Effects of moderate doses of ionizing radiation on experimental abdominal aortic aneurysm. PLoS One 2024; 19:e0308273. [PMID: 39088551 PMCID: PMC11293671 DOI: 10.1371/journal.pone.0308273] [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: 03/07/2024] [Accepted: 07/18/2024] [Indexed: 08/03/2024] Open
Abstract
BACKGROUND Exposure to ionizing radiation has been linked to cardiovascular diseases. However, the impact of moderate doses of radiation on abdominal aortic aneurysm (AAA) remains unknown. METHODS Angiotensin II-infused Apoe-/- mice were irradiated (acute, 1 Gray) either 3 days before (Day-3) or 1 day after (Day+1) pomp implantation. Isolated primary aortic vascular smooth muscle cells (VSMCs) were irradiated (acute 1 Gray) for mechanistic studies and functional testing in vitro. RESULTS Day-3 and Day+1 irradiation resulted in a significant reduction in aorta dilation (Control: 1.39+/-0.12; Day-3: 1.12+/-0.11; Day+1: 1.15+/-0.08 mm, P<0.001) and AAA incidence (Control: 81.0%; Day-3: 33.3%, Day+1: 53.3%) compared to the non-irradiated group. Day-3 and Day+1 irradiation led to an increase in collagen content in the adventitia (Thickness control: 23.64+/-2.9; Day-3: 54.39+/-15.5; Day+1 37.55+/-10.8 mm, P = 0.006). However, the underlying protective mechanisms were different between Day-3 and Day+1 groups. Irradiation before Angiotensin II (AngII) infusion mainly modulated vascular smooth muscle cell (VSMC) phenotype with a decrease in contractile profile and enhanced proliferative and migratory activity. Irradiation after AngII infusion led to an increase in macrophage content with a local anti-inflammatory phenotype characterized by the upregulation of M2-like gene and IL-10 expression. CONCLUSION Moderate doses of ionizing radiation mitigate AAA either through VSCM phenotype or inflammation modulation, depending on the time of irradiation.
Collapse
MESH Headings
- Animals
- Aortic Aneurysm, Abdominal/pathology
- Aortic Aneurysm, Abdominal/metabolism
- Aortic Aneurysm, Abdominal/etiology
- Mice
- Muscle, Smooth, Vascular/metabolism
- Muscle, Smooth, Vascular/radiation effects
- Muscle, Smooth, Vascular/pathology
- Radiation, Ionizing
- Angiotensin II/pharmacology
- Myocytes, Smooth Muscle/metabolism
- Myocytes, Smooth Muscle/radiation effects
- Myocytes, Smooth Muscle/pathology
- Male
- Disease Models, Animal
- Interleukin-10/metabolism
- Interleukin-10/genetics
- Collagen/metabolism
- Cell Proliferation/radiation effects
Collapse
Affiliation(s)
- Goran Riazi
- Experimental Radiotoxicology and Radiobiology Laboratory (LRTOX), Institute for Radiobiological Protection and Nuclear Safety (IRSN), Fontenay-aux-Roses, France
| | - Chloe Brizais
- Experimental Radiotoxicology and Radiobiology Laboratory (LRTOX), Institute for Radiobiological Protection and Nuclear Safety (IRSN), Fontenay-aux-Roses, France
| | - Imene Garali
- Experimental Radiotoxicology and Radiobiology Laboratory (LRTOX), Institute for Radiobiological Protection and Nuclear Safety (IRSN), Fontenay-aux-Roses, France
| | - Rida Al-rifai
- Université de Paris, Inserm U970, Paris-Cardiovascular Research Center, Paris, France
| | - Helene Quelquejay
- Experimental Radiotoxicology and Radiobiology Laboratory (LRTOX), Institute for Radiobiological Protection and Nuclear Safety (IRSN), Fontenay-aux-Roses, France
| | - Virginie Monceau
- Experimental Radiotoxicology and Radiobiology Laboratory (LRTOX), Institute for Radiobiological Protection and Nuclear Safety (IRSN), Fontenay-aux-Roses, France
| | - Guillaume Vares
- Experimental Radiotoxicology and Radiobiology Laboratory (LRTOX), Institute for Radiobiological Protection and Nuclear Safety (IRSN), Fontenay-aux-Roses, France
| | - Lea Ould-Boukhitine
- Experimental Radiotoxicology and Radiobiology Laboratory (LRTOX), Institute for Radiobiological Protection and Nuclear Safety (IRSN), Fontenay-aux-Roses, France
| | - Damien Aubeleau
- Experimental Radiotoxicology and Radiobiology Laboratory (LRTOX), Institute for Radiobiological Protection and Nuclear Safety (IRSN), Fontenay-aux-Roses, France
| | - Florian Gilain
- Experimental Radiotoxicology and Radiobiology Laboratory (LRTOX), Institute for Radiobiological Protection and Nuclear Safety (IRSN), Fontenay-aux-Roses, France
| | - Celine Gloaguen
- Experimental Radiotoxicology and Radiobiology Laboratory (LRTOX), Institute for Radiobiological Protection and Nuclear Safety (IRSN), Fontenay-aux-Roses, France
| | - Morgane Dos Santos
- Accidental Exposure Radiobiology Laboratory (LRACC), Institute for Radiobiological Protection and Nuclear Safety (IRSN), Fontenay-aux-Roses, France
| | - Hafid Ait-Oufella
- Université de Paris, Inserm U970, Paris-Cardiovascular Research Center, Paris, France
- Medical Intensive Care Unit, Hôpital Saint-Antoine, AP-HP, Sorbonne Université, Paris, France
| | - Teni Ebrahimian
- Experimental Radiotoxicology and Radiobiology Laboratory (LRTOX), Institute for Radiobiological Protection and Nuclear Safety (IRSN), Fontenay-aux-Roses, France
| |
Collapse
|
20
|
Huang X, Wang X, Wang Y, Shen S, Chen W, Liu T, Wang P, Fan X, Liu L, Jia J, Cong M. TIMP-1 Promotes Expression of MCP-1 and Macrophage Migration by Inducing Fli-1 in Experimental Liver Fibrosis. J Clin Transl Hepatol 2024; 12:634-645. [PMID: 38993513 PMCID: PMC11233975 DOI: 10.14218/jcth.2023.00514] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/14/2023] [Revised: 05/08/2024] [Accepted: 05/11/2024] [Indexed: 07/13/2024] Open
Abstract
Background and Aims Tissue inhibitor of metalloproteinase-1 (TIMP-1) plays a role in the excessive generation of extracellular matrix in liver fibrosis. This study aimed to explore the pathways through which TIMP-1 controls monocyte chemoattractant protein-1 (MCP-1) expression and promotes hepatic macrophage recruitment. Methods Liver fibrosis was triggered through carbon tetrachloride, and an adeno-associated virus containing small interfering RNA targeting TIMP-1 (siRNA-TIMP-1) was administered to both rats and mice. We assessed the extent of fibrosis and macrophage recruitment. The molecular mechanisms regulating macrophage recruitment by TIMP-1 were investigated through transwell migration assays, luciferase reporter assays, the use of pharmacological modulators, and an analysis of extracellular vesicles (EVs). Results siRNA-TIMP-1 alleviated carbon tetrachloride-induced liver fibrosis, reducing macrophage migration and MCP-1 expression. Co-culturing macrophages with hepatic stellate cells (HSCs) post-TIMP-1 downregulation inhibited macrophage migration. In siRNA-TIMP-1-treated HSCs, microRNA-145 (miRNA-145) expression increased, while the expression of Friend leukemia virus integration-1 (Fli-1) and MCP-1 was inhibited. Downregulation of Fli-1 led to decreased MCP-1 expression, whereas Fli-1 overexpression increased MCP-1 expression within HSCs. Transfection with miRNA-145 mimics reduced the expression of both Fli-1 and MCP-1, while miRNA-145 inhibitors elevated the expression of both Fli-1 and MCP-1 in HSCs. miRNA-145 bound directly to the 3'-UTR of Fli-1, and miRNA-145-enriched EVs secreted by HSCs after TIMP-1 downregulation influenced macrophage recruitment. Conclusions TIMP-1 induces Fli-1 expression through miRNA-145, subsequently increasing MCP-1 expression and macrophage recruitment. MiRNA-145-enriched EVs from HSCs can transmit biological information and magnify the function of TIMP-1.
Collapse
Affiliation(s)
- Xiaoli Huang
- Liver Research Center, Beijing Friendship Hospital, Capital Medical University, Beijing, China
- Beijing Key Laboratory of Translational Medicine in Liver Cirrhosis and National Clinical Research Center of Digestive Disease, Beijing, China
| | - Xiaofan Wang
- Liver Research Center, Beijing Friendship Hospital, Capital Medical University, Beijing, China
- Beijing Key Laboratory of Translational Medicine in Liver Cirrhosis and National Clinical Research Center of Digestive Disease, Beijing, China
| | - Yanhong Wang
- Liver Research Center, Beijing Friendship Hospital, Capital Medical University, Beijing, China
- Beijing Key Laboratory of Translational Medicine in Liver Cirrhosis and National Clinical Research Center of Digestive Disease, Beijing, China
- Dongying People's Hospital, Dongying, Shandong, China
| | - Shuangjun Shen
- Liver Research Center, Beijing Friendship Hospital, Capital Medical University, Beijing, China
- Beijing Key Laboratory of Translational Medicine in Liver Cirrhosis and National Clinical Research Center of Digestive Disease, Beijing, China
| | - Wei Chen
- Experimental and Translational Research Center, Beijing Friendship Hospital, Capital Medical University, Beijing, China
| | - Tianhui Liu
- Liver Research Center, Beijing Friendship Hospital, Capital Medical University, Beijing, China
- Beijing Key Laboratory of Translational Medicine in Liver Cirrhosis and National Clinical Research Center of Digestive Disease, Beijing, China
| | - Ping Wang
- Liver Research Center, Beijing Friendship Hospital, Capital Medical University, Beijing, China
- Beijing Key Laboratory of Translational Medicine in Liver Cirrhosis and National Clinical Research Center of Digestive Disease, Beijing, China
| | - Xu Fan
- Liver Research Center, Beijing Friendship Hospital, Capital Medical University, Beijing, China
- Beijing Key Laboratory of Translational Medicine in Liver Cirrhosis and National Clinical Research Center of Digestive Disease, Beijing, China
| | - Lin Liu
- Liver Research Center, Beijing Friendship Hospital, Capital Medical University, Beijing, China
- Beijing Key Laboratory of Translational Medicine in Liver Cirrhosis and National Clinical Research Center of Digestive Disease, Beijing, China
| | - Jidong Jia
- Liver Research Center, Beijing Friendship Hospital, Capital Medical University, Beijing, China
- Beijing Key Laboratory of Translational Medicine in Liver Cirrhosis and National Clinical Research Center of Digestive Disease, Beijing, China
| | - Min Cong
- Liver Research Center, Beijing Friendship Hospital, Capital Medical University, Beijing, China
- Beijing Key Laboratory of Translational Medicine in Liver Cirrhosis and National Clinical Research Center of Digestive Disease, Beijing, China
| |
Collapse
|
21
|
Shengxiao X, Xinxin S, Yunxiang Z, Zhijie T, Xiaofei T. Identification of a basement membrane-related gene signature for predicting prognosis, immune infiltration, and drug sensitivity in colorectal cancer. Front Oncol 2024; 14:1428176. [PMID: 39011483 PMCID: PMC11246870 DOI: 10.3389/fonc.2024.1428176] [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: 05/05/2024] [Accepted: 06/14/2024] [Indexed: 07/17/2024] Open
Abstract
Background Colorectal cancer (CRC) is the most common malignancy affecting the gastrointestinal tract. Extensive research indicates that basement membranes (BMs) may play a crucial role in the initiation and progression of the disease. Methods Data on the RNA expression patterns and clinicopathological information of patients with CRC were sourced from The Cancer Genome Atlas (TCGA) and the Gene Expression Omnibus (GEO) databases. A BM-linked risk signature for the prediction of overall survival (OS) was formulated using univariate Cox regression and combined machine learning techniques. Survival outcomes, functional pathways, the tumor microenvironment (TME), and responses to both immunotherapy and chemotherapy within varying risk classifications were also investigated. The expression trends of the model genes were evaluated by reverse transcription polymerase chain reaction (RT-PCR) and the Human Protein Atlas (HPA) database. Results A nine-gene risk signature containing UNC5C, TINAG, TIMP1, SPOCK3, MMP1, AGRN, UNC5A, ADAMTS4, and ITGA7 was constructed for the prediction of outcomes in patients with CRC. The expression profiles of these candidate genes were verified using RT-PCR and the HPA database and were found to be consistent with the findings on differential gene expression in the TCGA dataset. The validity of the signature was confirmed using the GEO cohort. The patients were stratified into different risk groups according to differences in clinicopathological characteristics, TME features, enrichment functions, and drug sensitivities. Lastly, the prognostic nomogram model based on the risk score was found to be effective in identifying high-risk patients and predicting OS. Conclusion A basement membrane-related risk signature was constructed and found to be effective for predicting the prognosis of patients with CRC.
Collapse
Affiliation(s)
- Xiang Shengxiao
- Department of Science and Education, Suqian First Hospital, Suqian, Jiangsu, China
| | - Sun Xinxin
- Department of Science and Education, Yangzhou Maternal and Child Health Hospital, Yangzhou, Jiangsu, China
| | - Zhu Yunxiang
- Department of Gastrointestinal Surgery, The Affiliated Hospital of Yangzhou University, Yangzhou University, Yangzhou, China
| | - Tang Zhijie
- Department of Gastrointestinal Surgery, The Affiliated Hospital of Yangzhou University, Yangzhou University, Yangzhou, China
| | - Tang Xiaofei
- Department of Gastrointestinal Surgery, The Affiliated Hospital of Yangzhou University, Yangzhou University, Yangzhou, China
| |
Collapse
|
22
|
Hellenbrand DJ, Quinn CM, Piper ZJ, Elder RT, Mishra RR, Marti TL, Omuro PM, Roddick RM, Lee JS, Murphy WL, Hanna AS. The secondary injury cascade after spinal cord injury: an analysis of local cytokine/chemokine regulation. Neural Regen Res 2024; 19:1308-1317. [PMID: 37905880 PMCID: PMC11467934 DOI: 10.4103/1673-5374.385849] [Citation(s) in RCA: 9] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2023] [Revised: 05/24/2023] [Accepted: 09/04/2023] [Indexed: 11/02/2023] Open
Abstract
After spinal cord injury, there is an extensive infiltration of immune cells, which exacerbates the injury and leads to further neural degeneration. Therefore, a major aim of current research involves targeting the immune response as a treatment for spinal cord injury. Although much research has been performed analyzing the complex inflammatory process following spinal cord injury, there remain major discrepancies within previous literature regarding the timeline of local cytokine regulation. The objectives of this study were to establish an overview of the timeline of cytokine regulation for 2 weeks after spinal cord injury, identify sexual dimorphisms in terms of cytokine levels, and determine local cytokines that significantly change based on the severity of spinal cord injury. Rats were inflicted with either a mild contusion, moderate contusion, severe contusion, or complete transection, 7 mm of spinal cord centered on the injury was harvested at varying times post-injury, and tissue homogenates were analyzed with a Cytokine/Chemokine 27-Plex assay. Results demonstrated pro-inflammatory cytokines including tumor necrosis factor α, interleukin-1β, and interleukin-6 were all upregulated after spinal cord injury, but returned to uninjured levels within approximately 24 hours post-injury, while chemokines including monocyte chemoattractant protein-1 remained upregulated for days post-injury. In contrast, several anti-inflammatory cytokines and growth factors including interleukin-10 and vascular endothelial growth factor were downregulated by 7 days post-injury. After spinal cord injury, tissue inhibitor of metalloproteinase-1, which specifically affects astrocytes involved in glial scar development, increased more than all other cytokines tested, reaching 26.9-fold higher than uninjured rats. After a mild injury, 11 cytokines demonstrated sexual dimorphisms; however, after a severe contusion only leptin levels were different between female and male rats. In conclusion, pro-inflammatory cytokines initiate the inflammatory process and return to baseline within hours post-injury, chemokines continue to recruit immune cells for days post-injury, while anti-inflammatory cytokines are downregulated by a week post-injury, and sexual dimorphisms observed after mild injury subsided with more severe injuries. Results from this work define critical chemokines that influence immune cell infiltration and important cytokines involved in glial scar development after spinal cord injury, which are essential for researchers developing treatments targeting secondary damage after spinal cord injury.
Collapse
Affiliation(s)
- Daniel J. Hellenbrand
- Department of Neurosurgery, University of Wisconsin-Madison School of Medicine and Public Health, Madison, WI, USA
- Department of Biomedical Engineering, University of Wisconsin-Madison, Madison, WI, USA
| | - Charles M. Quinn
- Department of Neurosurgery, University of Wisconsin-Madison School of Medicine and Public Health, Madison, WI, USA
| | - Zachariah J. Piper
- Department of Neurosurgery, University of Wisconsin-Madison School of Medicine and Public Health, Madison, WI, USA
| | - Ryan T. Elder
- Department of Neurosurgery, University of Wisconsin-Madison School of Medicine and Public Health, Madison, WI, USA
| | - Raveena R. Mishra
- Department of Neurosurgery, University of Wisconsin-Madison School of Medicine and Public Health, Madison, WI, USA
| | - Taylor L. Marti
- Department of Neurosurgery, University of Wisconsin-Madison School of Medicine and Public Health, Madison, WI, USA
| | - Phoebe M. Omuro
- Department of Neurosurgery, University of Wisconsin-Madison School of Medicine and Public Health, Madison, WI, USA
| | - Rylie M. Roddick
- Department of Neurosurgery, University of Wisconsin-Madison School of Medicine and Public Health, Madison, WI, USA
| | - Jae Sung Lee
- Department of Biomedical Engineering, University of Wisconsin-Madison, Madison, WI, USA
- Department of Orthopedics and Rehabilitation, University of Wisconsin-Madison, Madison, WI, USA
| | - William L. Murphy
- Department of Biomedical Engineering, University of Wisconsin-Madison, Madison, WI, USA
- Department of Orthopedics and Rehabilitation, University of Wisconsin-Madison, Madison, WI, USA
- Department of Materials Science and Engineering, University of Wisconsin-Madison, Madison, WI, USA
- Forward BIO Institute, University of Wisconsin-Madison, Madison, WI, USA
| | - Amgad S. Hanna
- Department of Neurosurgery, University of Wisconsin-Madison School of Medicine and Public Health, Madison, WI, USA
- Department of Biomedical Engineering, University of Wisconsin-Madison, Madison, WI, USA
| |
Collapse
|
23
|
Zhai J, Voraphani N, Imboden M, Keidel D, Liu C, Stern DA, Venker C, Petersen H, Bosco A, Sherrill DL, Morgan WJ, Tesfaigzi Y, Probst-Hensch NM, Martinez FD, Halonen M, Guerra S. Circulating biomarkers of airflow limitation across the life span. J Allergy Clin Immunol 2024; 153:1692-1703. [PMID: 38253260 PMCID: PMC11162345 DOI: 10.1016/j.jaci.2023.12.026] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2023] [Revised: 12/16/2023] [Accepted: 12/22/2023] [Indexed: 01/24/2024]
Abstract
BACKGROUND Airflow limitation is a hallmark of chronic obstructive pulmonary disease, which can develop through different lung function trajectories across the life span. There is a need for longitudinal studies aimed at identifying circulating biomarkers of airflow limitation across different stages of life. OBJECTIVES This study sought to identify a signature of serum proteins associated with airflow limitation and evaluate their relation to lung function longitudinally in adults and children. METHODS This study used data from 3 adult cohorts (TESAOD [Tucson Epidemiological Study of Airway Obstructive Disease], SAPALDIA [Swiss Cohort Study on Air Pollution and Lung and Heart Diseases in Adults], LSC [Lovelace Smoker Cohort]) and 1 birth cohort (TCRS [Tucson Children's Respiratory Study]) (N = 1940). In TESAOD, among 46 circulating proteins, we identified those associated with FEV1/forced vital capacity (FVC) percent (%) predicted levels and generated a score based on the sum of their z-scores. Cross-sectional analyses were used to test the score for association with concomitant lung function. Longitudinal analyses were used to test the score for association with subsequent lung function growth in childhood and decline in adult life. RESULTS After false discovery rate adjustment, serum levels of 5 proteins (HP, carcinoembryonic antigen, ICAM1, CRP, TIMP1) were associated with percent predicted levels of FEV1/FVC and FEV1 in TESAOD. In cross-sectional multivariate analyses the 5-biomarker score was associated with FEV1 % predicted in all adult cohorts (meta-analyzed FEV1 decrease for 1-SD score increase: -2.9%; 95% CI: -3.9%, -1.9%; P = 2.4 × 10-16). In multivariate longitudinal analyses, the biomarker score at 6 years of age was inversely associated with FEV1 and FEV1/FVC levels attained by young adult life (P = .02 and .005, respectively). In adults, persistently high levels of the biomarker score were associated with subsequent accelerated decline of FEV1 and FEV1/FVC (P = .01 and .001). CONCLUSIONS A signature of 5 circulating biomarkers of airflow limitation was associated with both impaired lung function growth in childhood and accelerated lung function decline in adult life, indicating that these proteins may be involved in multiple lung function trajectories leading to chronic obstructive pulmonary disease.
Collapse
Affiliation(s)
- Jing Zhai
- Asthma and Airway Disease Research Center, University of Arizona, Tucson, Ariz
| | - Nipasiri Voraphani
- Asthma and Airway Disease Research Center, University of Arizona, Tucson, Ariz
| | - Medea Imboden
- Swiss Tropical and Public Health Institute, Allschwil, Switzerland; University of Basel, Basel, Switzerland
| | - Dirk Keidel
- Swiss Tropical and Public Health Institute, Allschwil, Switzerland; University of Basel, Basel, Switzerland
| | - Congjian Liu
- Brigham and Women's Hospital, Harvard Medical School, Boston, Mass
| | - Debra A Stern
- Asthma and Airway Disease Research Center, University of Arizona, Tucson, Ariz
| | - Claire Venker
- Asthma and Airway Disease Research Center, University of Arizona, Tucson, Ariz
| | - Hans Petersen
- Lovelace Respiratory Research Institute, Albuquerque, NM
| | - Anthony Bosco
- Asthma and Airway Disease Research Center, University of Arizona, Tucson, Ariz
| | - Duane L Sherrill
- Asthma and Airway Disease Research Center, University of Arizona, Tucson, Ariz
| | - Wayne J Morgan
- Asthma and Airway Disease Research Center, University of Arizona, Tucson, Ariz
| | - Yohannes Tesfaigzi
- Brigham and Women's Hospital, Harvard Medical School, Boston, Mass; Lovelace Respiratory Research Institute, Albuquerque, NM
| | - Nicole M Probst-Hensch
- Swiss Tropical and Public Health Institute, Allschwil, Switzerland; University of Basel, Basel, Switzerland
| | - Fernando D Martinez
- Asthma and Airway Disease Research Center, University of Arizona, Tucson, Ariz
| | - Marilyn Halonen
- Asthma and Airway Disease Research Center, University of Arizona, Tucson, Ariz
| | - Stefano Guerra
- Asthma and Airway Disease Research Center, University of Arizona, Tucson, Ariz.
| |
Collapse
|
24
|
Iida K, Okada M. Identifying Key Regulatory Genes in Drug Resistance Acquisition: Modeling Pseudotime Trajectories of Breast Cancer Single-Cell Transcriptome. Cancers (Basel) 2024; 16:1884. [PMID: 38791962 PMCID: PMC11119661 DOI: 10.3390/cancers16101884] [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: 04/28/2024] [Revised: 05/11/2024] [Accepted: 05/15/2024] [Indexed: 05/26/2024] Open
Abstract
Single-cell RNA-sequencing (scRNA-seq) technology has provided significant insights into cancer drug resistance at the single-cell level. However, understanding dynamic cell transitions at the molecular systems level remains limited, requiring a systems biology approach. We present an approach that combines mathematical modeling with a pseudotime analysis using time-series scRNA-seq data obtained from the breast cancer cell line MCF-7 treated with tamoxifen. Our single-cell analysis identified five distinct subpopulations, including tamoxifen-sensitive and -resistant groups. Using a single-gene mathematical model, we discovered approximately 560-680 genes out of 6000 exhibiting multistable expression states in each subpopulation, including key estrogen-receptor-positive breast cancer cell survival genes, such as RPS6KB1. A bifurcation analysis elucidated their regulatory mechanisms, and we mapped these genes into a molecular network associated with cell survival and metastasis-related pathways. Our modeling approach comprehensively identifies key regulatory genes for drug resistance acquisition, enhancing our understanding of potential drug targets in breast cancer.
Collapse
Affiliation(s)
- Keita Iida
- Institute for Protein Research, Osaka University, Suita 565-0871, Osaka, Japan;
| | | |
Collapse
|
25
|
Tu Y, Jiang T, Wu Y, Luo J, Sun S, Liu C, Yu P, Chen A, Ji H, Wan Y, Yu L, Shi L. Difference of Antrochoanal Polyps Between Children and Adults in the Chinese Population. Laryngoscope 2024; 134:2093-2099. [PMID: 37916785 DOI: 10.1002/lary.31154] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2023] [Revised: 09/18/2023] [Accepted: 10/20/2023] [Indexed: 11/03/2023]
Abstract
OBJECTIVE This study aims to find the difference in clinical and immunopathological characteristics between children and adults with antrochoanal polyps (ACPs) in the Chinese population. METHODS The clinical data of 69 patients diagnosed with ACPs were retrospectively analyzed. Cytokine levels in 16 controls and 40 ACPs tissues were determined by quantitative real-time polymerase chain reaction (qPCR). The expression of matrix metalloproteinase (MMP)-9 was measured using qPCR, immunofluorescent staining, and western blot. RESULTS There were 51 (73.9%) children (<18 years old) and 18 (26.1%) adults (≥18 years old). The sex ratio differed significantly between the two groups (p = 0.0032). There were no significant differences in the nasal side of ACPs and approaches to surgery between the two groups. In both groups, the most common symptom was nasal obstruction, followed by nasal discharge. As for associated nasal diseases, there was a significant difference between the two groups in septal deviation (p = 0.0223). Adult patients showed significantly higher expression of IL-8 mRNA than children (p = 0.0424). The mRNA and protein levels of MMP-9 were also significantly higher in adult patients than in children (p = 0.0498 and 0.0009, respectively). CONCLUSION In the Chinese population, the comorbidities and immunopathological characteristics of adult ACP patients are different from those of children. The level of IL-8 and MMP-9 was significantly higher in ACPs of adults than in children, which may contribute to the more severe tissue remolding in adult ACP patients. LEVEL OF EVIDENCE 3 Laryngoscope, 134:2093-2099, 2024.
Collapse
Affiliation(s)
- Yanyi Tu
- Department of Otolaryngology-Head and Neck Surgery, Shandong Provincial ENT Hospital, Shandong University, Jinan, China
| | - Tianjiao Jiang
- Department of Otolaryngology-Head and Neck Surgery, Shandong Provincial ENT Hospital, Shandong University, Jinan, China
| | - Yisha Wu
- Department of Otolaryngology-Head and Neck Surgery, Shandong Provincial ENT Hospital, Shandong University, Jinan, China
- Department of Otolaryngology-Head and Neck Surgery, The Second Hospital, Shanxi Medical University, Taiyuan, China
| | - Jinfeng Luo
- Department of Otolaryngology-Head and Neck Surgery, Shandong Provincial ENT Hospital, Shandong University, Jinan, China
| | - Shujuan Sun
- Department of Otolaryngology-Head and Neck Surgery, Shandong Provincial ENT Hospital, Shandong University, Jinan, China
| | - Chuanping Liu
- Department of Otolaryngology-Head and Neck Surgery, Shandong Provincial ENT Hospital, Shandong University, Jinan, China
| | - Peng Yu
- Department of Otolaryngology-Head and Neck Surgery, Shandong Provincial ENT Hospital, Shandong University, Jinan, China
| | - Aiping Chen
- Department of Otolaryngology-Head and Neck Surgery, Shandong Provincial ENT Hospital, Shandong University, Jinan, China
| | - Hongzhi Ji
- Department of Otolaryngology-Head and Neck Surgery, Shandong Provincial ENT Hospital, Shandong University, Jinan, China
| | - Yuzhu Wan
- Department of Otolaryngology-Head and Neck Surgery, Shandong Provincial ENT Hospital, Shandong University, Jinan, China
| | - Liang Yu
- Department of Otolaryngology-Head and Neck Surgery, Shandong Provincial ENT Hospital, Shandong University, Jinan, China
| | - Li Shi
- Department of Otolaryngology-Head and Neck Surgery, Shandong Provincial ENT Hospital, Shandong University, Jinan, China
| |
Collapse
|
26
|
Duch P, Díaz‐Valdivia N, Gabasa M, Ikemori R, Arshakyan M, Fernández‐Nogueira P, Llorente A, Teixido C, Ramírez J, Pereda J, Chuliá‐Peris L, Galbis JM, Hilberg F, Reguart N, Radisky DC, Alcaraz J. Aberrant TIMP-1 production in tumor-associated fibroblasts drives the selective benefits of nintedanib in lung adenocarcinoma. Cancer Sci 2024; 115:1505-1519. [PMID: 38476010 PMCID: PMC11093210 DOI: 10.1111/cas.16141] [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: 10/03/2023] [Revised: 02/01/2024] [Accepted: 02/23/2024] [Indexed: 03/14/2024] Open
Abstract
The fibrotic tumor microenvironment is a pivotal therapeutic target. Nintedanib, a clinically approved multikinase antifibrotic inhibitor, is effective against lung adenocarcinoma (ADC) but not squamous cell carcinoma (SCC). Previous studies have implicated the secretome of tumor-associated fibroblasts (TAFs) in the selective effects of nintedanib in ADC, but the driving factor(s) remained unidentified. Here we examined the role of tissue inhibitor of metalloproteinase-1 (TIMP-1), a tumor-promoting cytokine overproduced in ADC-TAFs. To this aim, we combined genetic approaches with in vitro and in vivo preclinical models based on patient-derived TAFs. Nintedanib reduced TIMP-1 production more efficiently in ADC-TAFs than SCC-TAFs through a SMAD3-dependent mechanism. Cell culture experiments indicated that silencing TIMP1 in ADC-TAFs abolished the therapeutic effects of nintedanib on cancer cell growth and invasion, which were otherwise enhanced by the TAF secretome. Consistently, co-injecting ADC cells with TIMP1-knockdown ADC-TAFs into immunocompromised mice elicited a less effective reduction of tumor growth and invasion under nintedanib treatment compared to tumors bearing unmodified fibroblasts. Our results unveil a key mechanism underlying the selective mode of action of nintedanib in ADC based on the excessive production of TIMP-1 in ADC-TAFs. We further pinpoint reduced SMAD3 expression and consequent limited TIMP-1 production in SCC-TAFs as key for the resistance of SCC to nintedanib. These observations strongly support the emerging role of TIMP-1 as a critical regulator of therapy response in solid tumors.
Collapse
Affiliation(s)
- Paula Duch
- Department of Biomedicine, School of Medicine and Health Sciences, Unit of Biophysics and BioengineeringUniversity of BarcelonaBarcelonaSpain
| | - Natalia Díaz‐Valdivia
- Department of Biomedicine, School of Medicine and Health Sciences, Unit of Biophysics and BioengineeringUniversity of BarcelonaBarcelonaSpain
| | - Marta Gabasa
- Department of Biomedicine, School of Medicine and Health Sciences, Unit of Biophysics and BioengineeringUniversity of BarcelonaBarcelonaSpain
- Thoracic Oncology UnitHospital Clinic BarcelonaBarcelonaSpain
| | - Rafael Ikemori
- Department of Biomedicine, School of Medicine and Health Sciences, Unit of Biophysics and BioengineeringUniversity of BarcelonaBarcelonaSpain
| | - Marselina Arshakyan
- Department of Biomedicine, School of Medicine and Health Sciences, Unit of Biophysics and BioengineeringUniversity of BarcelonaBarcelonaSpain
| | - Patricia Fernández‐Nogueira
- Department of Biomedicine, School of Medicine and Health Sciences, Unit of Biophysics and BioengineeringUniversity of BarcelonaBarcelonaSpain
| | - Alejandro Llorente
- Department of Biomedicine, School of Medicine and Health Sciences, Unit of Biophysics and BioengineeringUniversity of BarcelonaBarcelonaSpain
| | - Cristina Teixido
- Thoracic Oncology UnitHospital Clinic BarcelonaBarcelonaSpain
- Pathology ServiceHospital Clinic BarcelonaBarcelonaSpain
- August Pi i Sunyer Biomedical Research Institute (IDIBAPS)BarcelonaSpain
| | - Josep Ramírez
- Thoracic Oncology UnitHospital Clinic BarcelonaBarcelonaSpain
- Pathology ServiceHospital Clinic BarcelonaBarcelonaSpain
- Biomedical Research Center Network for Respiratory Diseases (CIBERES)Carlos III Health InstituteMadridSpain
| | - Javier Pereda
- Department of Physiology, Faculty of PharmacyUniversity of ValenciaBurjassotSpain
| | - Lourdes Chuliá‐Peris
- Department of Physiology, Faculty of PharmacyUniversity of ValenciaBurjassotSpain
| | | | - Frank Hilberg
- Boehringer Ingelheim Austria RCV GmbH & Co. KGViennaAustria
| | - Noemí Reguart
- Thoracic Oncology UnitHospital Clinic BarcelonaBarcelonaSpain
- August Pi i Sunyer Biomedical Research Institute (IDIBAPS)BarcelonaSpain
| | | | - Jordi Alcaraz
- Department of Biomedicine, School of Medicine and Health Sciences, Unit of Biophysics and BioengineeringUniversity of BarcelonaBarcelonaSpain
- Thoracic Oncology UnitHospital Clinic BarcelonaBarcelonaSpain
- Biomedical Research Center Network for Respiratory Diseases (CIBERES)Carlos III Health InstituteMadridSpain
- Institute for Bioengineering of Catalonia (IBEC)The Barcelona Institute for Science and Technology (BIST)BarcelonaSpain
| |
Collapse
|
27
|
Zubkova E, Kalinin A, Bolotskaya A, Beloglazova I, Menshikov M. Autophagy-Dependent Secretion: Crosstalk between Autophagy and Exosome Biogenesis. Curr Issues Mol Biol 2024; 46:2209-2235. [PMID: 38534758 DOI: 10.3390/cimb46030142] [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: 11/30/2023] [Revised: 01/22/2024] [Accepted: 01/23/2024] [Indexed: 03/28/2024] Open
Abstract
The cellular secretome is pivotal in mediating intercellular communication and coordinating responses to stressors. Exosomes, initially recognized for their role in waste disposal, have now emerged as key intercellular messengers with significant therapeutic and diagnostic potential. Similarly, autophagy has transcended its traditional role as a waste removal mechanism, emerging as a regulator of intracellular communication pathways and a contributor to a unique autophagy-dependent secretome. Secretory authophagy, initiated by various stress stimuli, prompts the selective release of proteins implicated in inflammation, including leaderless proteins that bypass the conventional endoplasmic reticulum-Golgi secretory pathway. This reflects the significant impact of stress-induced autophagy on cellular secretion profiles, including the modulation of exosome release. The convergence of exosome biogenesis and autophagy is exemplified by the formation of amphisomes, vesicles that integrate autophagic and endosomal pathways, indicating their synergistic interplay. Regulatory proteins common to both pathways, particularly mTORC1, emerge as potential therapeutic targets to alter cellular secretion profiles involved in various diseases. This review explores the dynamic interplay between autophagy and exosome formation, highlighting the potential to influence the secretome composition. While the modulation of exosome secretion and cytokine preconditioning is well-established in regenerative medicine, the strategic manipulation of autophagy is still underexplored, presenting a promising but uncharted therapeutic landscape.
Collapse
Affiliation(s)
- Ekaterina Zubkova
- National Medical Research Centre of Cardiology Named after Academician E.I. Chazov, 121552 Moscow, Russia
| | - Alexander Kalinin
- National Medical Research Centre of Cardiology Named after Academician E.I. Chazov, 121552 Moscow, Russia
- Faculty of Fundamental Medicine, Lomonosov Moscow State University, 119991 Moscow, Russia
| | - Anastasya Bolotskaya
- National Medical Research Centre of Cardiology Named after Academician E.I. Chazov, 121552 Moscow, Russia
- Institute of Clinical Medicine, Sechenov University, 119435 Moscow, Russia
| | - Irina Beloglazova
- National Medical Research Centre of Cardiology Named after Academician E.I. Chazov, 121552 Moscow, Russia
| | - Mikhail Menshikov
- National Medical Research Centre of Cardiology Named after Academician E.I. Chazov, 121552 Moscow, Russia
| |
Collapse
|
28
|
Coates-Park S, Rich JA, Stetler-Stevenson WG, Peeney D. The TIMP protein family: diverse roles in pathophysiology. Am J Physiol Cell Physiol 2024; 326:C917-C934. [PMID: 38284123 PMCID: PMC11193487 DOI: 10.1152/ajpcell.00699.2023] [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: 12/28/2023] [Revised: 01/23/2024] [Accepted: 01/23/2024] [Indexed: 01/30/2024]
Abstract
The tissue inhibitors of matrix metalloproteinases (TIMPs) are a family of four matrisome proteins classically defined by their roles as the primary endogenous inhibitors of metalloproteinases (MPs). Their functions however are not limited to MP inhibition, with each family member harboring numerous MP-independent biological functions that play key roles in processes such as inflammation and apoptosis. Because of these multifaceted functions, TIMPs have been cited in diverse pathophysiological contexts. Herein, we provide a comprehensive overview of the MP-dependent and -independent roles of TIMPs across a range of pathological conditions. The potential therapeutic and biomarker applications of TIMPs in these disease contexts are also considered, highlighting the biomedical promise of this complex and often misunderstood protein family.
Collapse
Affiliation(s)
- Sasha Coates-Park
- Extracellular Matrix Pathology Section, Laboratory of Pathology, National Cancer Institute, National Institute of Health, Bethesda, Maryland, United States
| | - Joshua A Rich
- Extracellular Matrix Pathology Section, Laboratory of Pathology, National Cancer Institute, National Institute of Health, Bethesda, Maryland, United States
| | - William G Stetler-Stevenson
- Extracellular Matrix Pathology Section, Laboratory of Pathology, National Cancer Institute, National Institute of Health, Bethesda, Maryland, United States
| | - David Peeney
- Extracellular Matrix Pathology Section, Laboratory of Pathology, National Cancer Institute, National Institute of Health, Bethesda, Maryland, United States
| |
Collapse
|
29
|
Nowak-Wąs M, Wąs P, Czuba Z, Wojnicz R, Wyględowska-Promieńska D. Expression of Tissue Inhibitors of Metalloproteinases (TIMP-1, TIMP-2, TIMP-3, TIMP-4) in Blood Serum of Patients with Keratoconus. J Clin Med 2024; 13:1168. [PMID: 38398480 PMCID: PMC10889408 DOI: 10.3390/jcm13041168] [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: 01/19/2024] [Revised: 02/13/2024] [Accepted: 02/16/2024] [Indexed: 02/25/2024] Open
Abstract
BACKGROUND The etiology of keratoconus is unclear. Current evidence suggests that inflammatory and systemic mechanisms might play a role in its pathophysiology. The proper interaction of proteolytic enzymes-matrix metalloproteinases-and their specific tissue inhibitors (TIMPs) within the cornea is essential in maintaining its structure, transparency and healing processes. The aim of the study was to determine the concentration of the TIMPs TIMP-1, TIMP-2, TIMP-3, and TIMP-4 in the blood serum samples of patients with keratoconus compared to the control group. METHODS The study encompassed 132 patients, of which 83 people constituted the study group and 49 the control group. The concentration of selected TIMPs was determined using the Human Magnetic Luminex® Performance Assay method. RESULTS In the study group, the concentrations of TIMP-1 and TIMP-3 were statistically significantly reduced, and TIMP-2 and TIMP-4 increased compared to the control group. The analysis of individual TIMPs in terms of their usefulness as potential predictors of keratoconus showed high results of diagnostic sensitivity and specificity for all TIMPs, in particular for TIMP-1 and TIMP-2. CONCLUSION The above results may indicate systemic disturbances in the TIMPs regulation among keratoconus patients. High diagnostic sensitivity and specificity of all TIMPs, in particular TIMP-1 and TIMP-2, may confirm their participation in the etiopathogenesis of this disease.
Collapse
Affiliation(s)
- Marta Nowak-Wąs
- Department of Histology and Cell Pathology, Faculty of Medical Sciences in Zabrze, Medical University of Silesia, 40-055 Katowice, Poland
- Department of Ophthalmology, Kornel Gibinski University Clinical Center, Medical University of Silesia, 40-055 Katowice, Poland
| | - Paweł Wąs
- Department of Ophthalmology, Megrez Provincial Specialist Hospital in Tychy, 43-100 Tychy, Poland
| | - Zenon Czuba
- Department of Microbiology and Immunology, Faculty of Medical Sciences in Zabrze, Medical University of Silesia, 40-055 Katowice, Poland
| | - Romuald Wojnicz
- Department of Histology and Cell Pathology, Faculty of Medical Sciences in Zabrze, Medical University of Silesia, 40-055 Katowice, Poland
| | - Dorota Wyględowska-Promieńska
- Department of Ophthalmology, Kornel Gibinski University Clinical Center, Medical University of Silesia, 40-055 Katowice, Poland
- Department of Ophthalmology, Faculty of Medical Sciences in Katowice, Medical University of Silesia, 40-055 Katowice, Poland
| |
Collapse
|
30
|
Sampaio Moura N, Schledwitz A, Alizadeh M, Patil SA, Raufman JP. Matrix metalloproteinases as biomarkers and therapeutic targets in colitis-associated cancer. Front Oncol 2024; 13:1325095. [PMID: 38288108 PMCID: PMC10824561 DOI: 10.3389/fonc.2023.1325095] [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: 10/20/2023] [Accepted: 12/26/2023] [Indexed: 01/31/2024] Open
Abstract
Colorectal cancer (CRC) remains a major cause of morbidity and mortality. Therapeutic approaches for advanced CRC are limited and rarely provide long-term benefit. Enzymes comprising the 24-member matrix metalloproteinase (MMP) family of zinc- and calcium-dependent endopeptidases are key players in extracellular matrix degradation, a requirement for colon tumor expansion, invasion, and metastasis; hence, MMPs are an important research focus. Compared to sporadic CRC, less is known regarding the molecular mechanisms and the role of MMPs in the development and progression of colitis-associated cancer (CAC) - CRC on a background of chronic inflammatory bowel disease (IBD) - primarily ulcerative colitis and Crohn's disease. Hence, the potential of MMPs as biomarkers and therapeutic targets for CAC is uncertain. Our goal was to review data regarding the role of MMPs in the development and progression of CAC. We sought to identify promising prognostic and therapeutic opportunities and novel lines of investigation. A key observation is that since MMPs may be more active in early phases of CAC, using MMPs as biomarkers of advancing neoplasia and as potential therapeutic targets for adjuvant therapy in those with advanced stage primary CAC rather than overt metastases may yield more favorable outcomes.
Collapse
Affiliation(s)
- Natalia Sampaio Moura
- Department of Medicine, Division of Gastroenterology and Hepatology, University of Maryland School of Medicine, Baltimore, MD, United States
| | - Alyssa Schledwitz
- Department of Medicine, Division of Gastroenterology and Hepatology, University of Maryland School of Medicine, Baltimore, MD, United States
| | - Madeline Alizadeh
- The Institute for Genome Sciences, University of Maryland School of Medicine, Baltimore, MD, United States
| | - Seema A. Patil
- Department of Medicine, Division of Gastroenterology and Hepatology, University of Maryland School of Medicine, Baltimore, MD, United States
| | - Jean-Pierre Raufman
- Department of Medicine, Division of Gastroenterology and Hepatology, University of Maryland School of Medicine, Baltimore, MD, United States
- Medical Service, Veterans Affairs Maryland Healthcare System, Baltimore, MD, United States
- Marlene and Stewart Greenebaum Cancer Center, University of Maryland Medical Center, Baltimore, MD, United States
- Department of Biochemistry and Molecular Biology, University of Maryland School of Medicine, Baltimore, MD, United States
| |
Collapse
|
31
|
Li M, Ni QY, Yu SY. Integration of single-cell transcriptomics and epigenetic analysis reveals enhancer-controlled TIMP1 as a regulator of ferroptosis in colorectal cancer. Genes Genomics 2024; 46:121-133. [PMID: 38032469 DOI: 10.1007/s13258-023-01474-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2023] [Accepted: 10/26/2023] [Indexed: 12/01/2023]
Abstract
BACKGROUND Ferroptosis is an iron-dependent non-apoptotic programmed cell death. However, the regulatory mechanism of ferroptosis in colorectal cancer (CRC) is still unclear. OBJECTIVE The aim of this study was to investigate the role and mechanism of enhancer-controlled genes in ferroptosis in CRC. METHODS Dimensionality reduction and differentially expressed genes (DEGs) identification were conducted using Seurat algorithm based on single-cell RNA sequencing (scRNA-seq) data from the GSE200997 dataset. Ferroptosis-related pathway enrichment analysis was performed using the FerrDb V2 database. Enhancers were identified using HOMER algorithm based on H3K27ac ChIP-seq data from the GSE166254 dataset. Kaplan-Meier Plotter online tool was used to analyze prognosis and gene expression correlation. Transcription factors were predicted using the transcription factor affinity prediction web tool. The binding of enhancer to transcription factor and H3K27ac enrichment were detected by ChIP-qPCR. RSL3 was used to induce ferroptosis in CRC cells. Gene transcription was detected by qRT-PCR. Cell proliferation was detected by CCK8 assay. RESULTS Nine cell clusters including T cells, natural killer cells, macrophages, mast cells, epithelial cells, fibroblasts, goblet cells, B cells and dendritic cells were identified in CRC and normal colonic tissue samples. Compared to normal colonic tissue-derived epithelial cells, 1075 DEGs were screened in CRC tissue-derived epithelial cells. Ferroptosis-related pathway enrichment suggested that DEGs were associated with the regulation of ferroptosis. DPEP1, ETV4, CEBPG, TIMP1, DUOX2 and LCN2 were identified as the significantly upregulated genes enriched in the "ferroptosis regulator" term, and their H3K27ac signals were significantly higher in CRC tissues than in normal colonic tissues. Of these, only the expression of TIMP1 predicted a poor prognosis of CRC patients. Transcription factor SPI1 drove TIMP1 transcription by binding to its enhancer. Overexpression of TIMP1 significantly promoted the resistance to ferroptosis induced by RSL3 in CRC cells, which was partially restored by SPI1 knockdown. CONCLUSION Transcription of TIMP1 was driven by transcription factor SPI1 in combination with its enhancer, consequently promoting CRC cells against ferroptosis. The SPI1/TIMP1 axis confers ferroptosis resistance in CRC, and thus has the potential to be the molecular targets for CRC treatment.
Collapse
Affiliation(s)
- Meng Li
- Department of Gastrointestinal Surgery, Hebei Medical University Third Hospital, Shijiazhuang, Hebei, 050051, China
| | - Qian-Yang Ni
- Department of Gastrointestinal Surgery, Hebei Medical University Third Hospital, Shijiazhuang, Hebei, 050051, China
| | - Su-Yang Yu
- Department of Gastrointestinal Surgery, Hebei Medical University Third Hospital, Shijiazhuang, Hebei, 050051, China.
| |
Collapse
|
32
|
Konrad ER, Soo J, Conroy AL, Namasopo S, Opoka RO, Hawkes MT. Circulating markers of neutrophil activation and lung injury in pediatric pneumonia in low-resource settings. Pathog Glob Health 2023; 117:708-716. [PMID: 36562081 PMCID: PMC10614712 DOI: 10.1080/20477724.2022.2160885] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022] Open
Abstract
Diagnostic biomarkers for childhood pneumonia could guide management and improve antibiotic stewardship in low-resource settings where chest x-ray (CXR) is not always available. In this cross-sectional study, we measured chitinase 3-like protein 1 (CHI3L1), surfactant protein D (SP-D), lipocalin-2 (LCN2), and tissue inhibitor of metalloproteinases-1 (TIMP-1) in Ugandan children under the age of five hospitalized with acute lower respiratory tract infection. We determined the association between biomarker levels and primary end-point pneumonia, indicated by CXR consolidation. We included 89 children (median age 11 months, 39% female). Primary endpoint pneumonia was present in 22 (25%). Clinical signs were similar in children with and without CXR consolidation. Broad-spectrum antibiotics (ceftriaxone) were administered in 83 (93%). Levels of CHI3L1, SP-D, LCN2 and TIMP-1 were higher in patients with primary end-point pneumonia compared to patients with normal CXR or other infiltrates. All markers were moderately accurate predictors of primary end-point pneumonia, with area under receiver operator characteristic curves of 0.66-0.70 (p<0.05 for all markers). The probability of CXR consolidation increased monotonically with the number of markers above cut-off. Among 28 patients (31%) in whom all four markers were below the cut-off, the likelihood ratio of CXR consolidation was 0.11 (95%CI 0.015 to 0.73). CHI3L1, SP-D, LCN2 and TIMP-1 were associated with CXR consolidation in children with clinical pneumonia in a low-resource setting. Combinations of quantitative biomarkers may be useful to safely withhold antibiotics in children with a low probability of bacterial infection.
Collapse
Affiliation(s)
- Emily R. Konrad
- Department of Pediatrics, University of Alberta, Edmonton, Canada
| | - Jeremy Soo
- Department of Pediatrics, University of Alberta, Edmonton, Canada
| | - Andrea L. Conroy
- Ryan White Center for Pediatric Infectious Diseases and Global Health, Indiana University School of Medicine, Indianapolis, USA
| | - Sophie Namasopo
- Department of Pediatrics, Kabale District Hospital, Kabale, Uganda
| | - Robert O. Opoka
- Department of Paediatrics and Child Health, Mulago Hospital and Makerere University, Kampala, Uganda
| | - Michael T. Hawkes
- Department of Pediatrics, University of Alberta, Edmonton, Canada
- School of Public Health, University of Alberta, Edmonton, Canada
- Department of Medical Microbiology and Immunology, University of Alberta, Edmonton, Canada
- Distinguished Researcher, Stollery Science Lab, Edmonton, Canada
- Member, Women and Children’s Health Research Institute, Edmonton, Canada
| |
Collapse
|
33
|
Abdolahi F, Shahraki A, Sheervalilou R, Mortazavi SS. Identification of differentially expressed genes associated with the pathogenesis of gastric cancer by bioinformatics analysis. BMC Med Genomics 2023; 16:311. [PMID: 38041130 PMCID: PMC10690994 DOI: 10.1186/s12920-023-01720-7] [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: 05/28/2023] [Accepted: 10/29/2023] [Indexed: 12/03/2023] Open
Abstract
AIM Gastric cancer (GC) is one of the most diagnosed cancers worldwide. GC is a heterogeneous disease whose pathogenesis has not been entirely understood. Besides, the GC prognosis for patients remains poor. Hence, finding reliable biomarkers and therapeutic targets for GC patients is urgently needed. METHODS GSE54129 and GSE26942 datasets were downloaded from Gene Expression Omnibus (GEO) database to detect differentially expressed genes (DEGs). Then, gene set enrichment analyses and protein-protein interactions were investigated. Afterward, ten hub genes were identified from the constructed network of DEGs. Then, the expression of hub genes in GC was validated. Performing survival analysis, the prognostic value of each hub gene in GC samples was investigated. Finally, the databases were used to predict microRNAs that could regulate the hub genes. Eventually, top miRNAs with more interactions with the list of hub genes were introduced. RESULTS In total, 203 overlapping DEGs were identified between both datasets. The main enriched KEGG pathway was "Protein digestion and absorption." The most significant identified GO terms included "primary alcohol metabolic process," "basal part of cell," and "extracellular matrix structural constituent conferring tensile strength." Identified hub modules were COL1A1, COL1A2, TIMP1, SPP1, COL5A2, THBS2, COL4A1, MUC6, CXCL8, and BGN. The overexpression of seven hub genes was associated with overall survival. Moreover, among the list of selected miRNAs, hsa-miR-27a-3, hsa-miR-941, hsa-miR-129-2-3p, and hsa-miR-1-3p, were introduced as top miRNAs targeting more than five hub genes. CONCLUSIONS The present study identified ten genes associated with GC, which may help discover novel prognostic and diagnostic biomarkers as well as therapeutic targets for GC. Our results may advance the understanding of GC occurrence and progression.
Collapse
Affiliation(s)
- Fatemeh Abdolahi
- Department of Biology, Faculty of Science, University of Sistan and Baluchestan, Zahedan, Iran
| | - Ali Shahraki
- Department of Biology, Faculty of Science, University of Sistan and Baluchestan, Zahedan, Iran
| | - Roghayeh Sheervalilou
- Pharmacology Research Center, Zahedan University of Medical Sciences, Zahedan, Iran.
| | | |
Collapse
|
34
|
Kansal H, Chopra V, Garg K, Sharma S. Role of thioredoxin in chronic obstructive pulmonary disease (COPD): a promising future target. Respir Res 2023; 24:295. [PMID: 38001457 PMCID: PMC10668376 DOI: 10.1186/s12931-023-02574-4] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2023] [Accepted: 10/22/2023] [Indexed: 11/26/2023] Open
Abstract
INTRODUCTION Thioredoxin (Trx) is a secretory protein that acts as an antioxidant, redox regulator, anti-allergic, and anti-inflammatory molecule. It has been used to treat dermatitis and inflammation of the digestive tract. In the lungs, Trx has a significant anti-inflammatory impact. On the other hand, Chronic Obstructive Pulmonary Disease (COPD) is one of the significant causes of death in the developed world, with a tremendous individual and socioeconomic impact. Despite new initiatives and endless treatment trials, COPD incidence and death will likely escalate in the coming decades. AREAS COVERED COPD is a chronic inflammatory disease impacting the airways, lung parenchyma, and pulmonary vasculature. Oxidative stress and protease-antiprotease imbalances are thought to be involved in the process. The most popular respiratory inflammatory and allergic disorders therapies are corticosteroids and β-receptor agonists. These medications are helpful but have some drawbacks, such as infection and immunosuppression; thus, addressing Trx signalling treatments may be a viable COPD treatment approach. This review shall cover the pathophysiology of COPD, the pharmacognosy of anti-COPD drugs, including the assets and liabilities of each, and the role and mechanism of Trx in COPD treatment. EXPERT OPINION Limited research has targeted the thioredoxin system as an anti-COPD drug. Spectating the increase in the mortality rates of COPD, this review article would be an interesting one to research.
Collapse
Affiliation(s)
- Heena Kansal
- Department of Biotechnology, Thapar Institute of Engineering and Technology, Patiala, India
| | - Vishal Chopra
- Department of Pulmonary Medicine, Government Medical College, Patiala, India
| | - Kranti Garg
- Department of Pulmonary Medicine, Government Medical College, Patiala, India
| | - Siddharth Sharma
- Department of Biotechnology, Thapar Institute of Engineering and Technology, Patiala, India.
| |
Collapse
|
35
|
Accolla RP, Deller M, Lansberry TR, Simmons A, Liang JP, Patel SN, Jiang K, Stabler CL. 3D printed elastomeric biomaterial mitigates compaction during in vitro vasculogenesis. Acta Biomater 2023; 171:363-377. [PMID: 37739251 PMCID: PMC11146342 DOI: 10.1016/j.actbio.2023.09.026] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2023] [Revised: 09/11/2023] [Accepted: 09/15/2023] [Indexed: 09/24/2023]
Abstract
A key parameter for the success of most cellular implants is the formation of a complete and comprehensive intra-implant vessel network. Pre-vascularization, the generation of vessel structures in vitro prior to transplantation, provides accelerated implant perfusion via anastomosis, but scalability and ease of integration hinder clinical translation. For fibrin-based vasculogenesis approaches, the remodeling and degradation of the fragile, hydrogel matrix during the formation of vessel-like structures results in rapid, cell-mediated construct compaction leading to dense, capillary-like structures with ineffective network coverage. To resolve these challenges, vasculogenic hydrogels were embedded within a highly porous, biostable three-dimensional (3D) polydimethylsiloxane (PDMS) scaffold. Using reverse-casting of 3D-printed molds, scaffolds exhibited highly interconnected and reproducible pore structures. Pore size was optimized via in vivo screening of intra-device angiogenesis. The inclusion of the PDMS frame with vasculogenic hydrogels significantly reduced fibrin compaction in vitro, resulting in easily manipulated constructs with predictable dimensionality and increased surface area compared to fibrin hydrogel alone. Globally, vascular morphogenesis was altered by the PDMS frame, with significantly larger and less dense network structures. Vasculogenic proteomic evaluation showed a temporal impact of the addition of the PDMS frame, indicating altered cellular proliferation and migration signaling. This work establishes a platform for improving the generation of translational pre-vascularized networks for greater flexibility to meet the needs of clinically scaled, engineered tissues. STATEMENT OF SIGNIFICANCE: Competent intra-implant vascularization is a significant issue hindering the success of engineered tissues. Pre-vascularization approaches, whereby a vascular network is formed in vitro and subsequently implanted into the host to anastomose, is a promising approach but it is limited by the compacted, dense, and poorly functional microcapillary structures typically formed using soft hydrogels. Herein, we have uniquely addressed this challenge by adding a 3D printed PDMS-based open framework structure that serves to prevent hydrogel compaction. Globally, we observed distinct differences in overall construct geometry, vascular network density, compaction, and morphogenesis, indicating that this PDMS framework lead to elevated maturity of this in vitro network while retaining its global dimensions. Overall, this novel approach elevates the translational potential of pre-vascularized constructs.
Collapse
Affiliation(s)
- Robert P Accolla
- J. Crayton Pruitt Family Department of Biomedical Engineering, University of Florida, Gainesville, FL, USA
| | - Madison Deller
- J. Crayton Pruitt Family Department of Biomedical Engineering, University of Florida, Gainesville, FL, USA
| | - Taylor R Lansberry
- J. Crayton Pruitt Family Department of Biomedical Engineering, University of Florida, Gainesville, FL, USA
| | - Amberlyn Simmons
- J. Crayton Pruitt Family Department of Biomedical Engineering, University of Florida, Gainesville, FL, USA
| | - Jia-Pu Liang
- J. Crayton Pruitt Family Department of Biomedical Engineering, University of Florida, Gainesville, FL, USA
| | - Smit N Patel
- J. Crayton Pruitt Family Department of Biomedical Engineering, University of Florida, Gainesville, FL, USA
| | - Kaiyuan Jiang
- J. Crayton Pruitt Family Department of Biomedical Engineering, University of Florida, Gainesville, FL, USA
| | - Cherie L Stabler
- J. Crayton Pruitt Family Department of Biomedical Engineering, University of Florida, Gainesville, FL, USA; Department of Immunology and Pathology, College of Medicine, University of Florida, Gainesville, FL, USA; University of Florida Diabetes Institute, Gainesville, FL, USA.
| |
Collapse
|
36
|
Manríquez-Treviño Y, Sánchez-Ramírez B, Grado-Ahuir JA, Castro-Valenzuela B, González-Horta C, Burrola-Barraza M. Human TIMP1 Is a Growth Factor That Improves Oocyte Developmental Competence. BIOTECH 2023; 12:60. [PMID: 37873882 PMCID: PMC10594479 DOI: 10.3390/biotech12040060] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2023] [Revised: 08/24/2023] [Accepted: 10/05/2023] [Indexed: 10/25/2023] Open
Abstract
Oocyte developmental competence is the ability of a mature oocyte to be fertilized and subsequently support embryonic development. Such competence is gained during folliculogenesis and is facilitated by the bidirectional communication into a compacted cumulus-oocyte complex (COC). Human tissue inhibitor of metalloproteinases-1 (TIMP1) participates in biological processes, including cell growth, differentiation, and apoptosis. This study aimed to evaluate the influence of TIMP1 as a growth factor on the in vitro maturation (IVM) culture of bovine COCs to improve oocyte developmental competence. All TIMP1 treatments (50, 100, and 150 ng/mL) favored the COCs' compaction structure (p < 0.05). TIMP1 at 150 ng/mL produced more oocytes in metaphase II compared to the other treatments (p < 0.05). The 150 ng/mL TIMP1 generated oocytes with the most (p < 0.05) cortical granules below the plasma membrane (pattern I). In a parthenogenesis assay, oocyte IVM in 50 ng/mL of TIMP1 produced the most blastocyst compared to the other treatments (p < 0.05). The Principal Component Analysis (PCA) showed that 50 ng/mL of TIMP1 was the best condition to develop oocyte competence because it was associated with the COC compact and cortical granule pattern I. TIMP1 influences the development of oocyte competence when added to the IVM culture medium of COCs.
Collapse
Affiliation(s)
- Yolanda Manríquez-Treviño
- Facultad de Zootecnia y Ecología, Universidad Autónoma de Chihuahua (UACH), Perif. Fco. R. Almada Km. 1, Chihuahua 31453, Chihuahua, Mexico; (Y.M.-T.); (J.A.G.-A.); (B.C.-V.)
| | - Blanca Sánchez-Ramírez
- Facultad de Ciencias Químicas, Universidad Autónoma de Chihuahua (UACH), Campus Universitario #2, Chihuahua 31125, Chihuahua, Mexico; (B.S.-R.); (C.G.-H.)
| | - Juan Alberto Grado-Ahuir
- Facultad de Zootecnia y Ecología, Universidad Autónoma de Chihuahua (UACH), Perif. Fco. R. Almada Km. 1, Chihuahua 31453, Chihuahua, Mexico; (Y.M.-T.); (J.A.G.-A.); (B.C.-V.)
| | - Beatriz Castro-Valenzuela
- Facultad de Zootecnia y Ecología, Universidad Autónoma de Chihuahua (UACH), Perif. Fco. R. Almada Km. 1, Chihuahua 31453, Chihuahua, Mexico; (Y.M.-T.); (J.A.G.-A.); (B.C.-V.)
| | - Carmen González-Horta
- Facultad de Ciencias Químicas, Universidad Autónoma de Chihuahua (UACH), Campus Universitario #2, Chihuahua 31125, Chihuahua, Mexico; (B.S.-R.); (C.G.-H.)
| | - M.Eduviges Burrola-Barraza
- Facultad de Zootecnia y Ecología, Universidad Autónoma de Chihuahua (UACH), Perif. Fco. R. Almada Km. 1, Chihuahua 31453, Chihuahua, Mexico; (Y.M.-T.); (J.A.G.-A.); (B.C.-V.)
| |
Collapse
|
37
|
Bąkowski P, Mieloch AA, Porzucek F, Mańkowska M, Ciemieniewska-Gorzela K, Naczk J, Piontek T, Rybka JD. Meniscus repair via collagen matrix wrapping and bone marrow injection: clinical and biomolecular study. INTERNATIONAL ORTHOPAEDICS 2023; 47:2409-2417. [PMID: 36764942 PMCID: PMC10522727 DOI: 10.1007/s00264-023-05711-2] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/02/2022] [Accepted: 01/22/2023] [Indexed: 02/12/2023]
Abstract
PURPOSE The functional outcomes of arthroscopic matrix-based meniscus repair (AMMR) in patients two and five years after the treatment clearly show that the use of the collagen matrix and bone marrow aspirate creates favorable biological conditions for meniscus healing. This study not only provides ten follow-up results but also investigates biomolecular mechanisms governing the regenerative process. METHODS Case series was based on data collected from patients who underwent AMMR procedure, starting with preoperatively through two-year and five-year till ten-year follow-up. The outcomes are presented as IKDC and the Lysholm subjective scores as well as the imaging results. Biomolecular investigation of the membranes utilized in the AMMR procedure include DNA content analysis, cell viability and proliferation study of bone marrow and bone marrow concentrate-derived cells, and cytokine array performed on monocytes cultured on the membranes. CONCLUSION Data collected from patients who underwent AMMR procedure, starting with pre-operatively through two year and five year till ten year follow-up, indicate the possibility for long-term, stable meniscus preservation. Outcomes are manifested with a visible improvement of the IKDC and the Lysholm subjective scores as well as in the imaging results. The type of the meniscal tear or complexity of the knee injury (isolated AMMR vs. AMMR + ACL) did not affect the clinical outcomes. The study highlighted the role of the membrane in facilitating cell adhesion and proliferation. Additionally, several cytokines were selected as potentially crucial products of the membrane vs. monocyte interactions, driving the tissue regeneration and remodeling. Interestingly, thresholds of what constitutes a safe and well-decellularized membrane according to relevant literature have been significantly breached, but ultimately did not elicit detrimental side effects.
Collapse
Affiliation(s)
- Paweł Bąkowski
- Department of Orthopedic Surgery, Rehasport Clinic, Poznan, Poland
| | - Adam Aron Mieloch
- Center for Advanced Technology, Adam Mickiewicz University in Poznan, Poznan, Poland
| | - Filip Porzucek
- Center for Advanced Technology, Adam Mickiewicz University in Poznan, Poznan, Poland
| | - Monika Mańkowska
- Center for Advanced Technology, Adam Mickiewicz University in Poznan, Poznan, Poland
| | | | - Jakub Naczk
- Department of Orthopedic Surgery, Rehasport Clinic, Poznan, Poland
| | - Tomasz Piontek
- Department of Orthopedic Surgery, Rehasport Clinic, Poznan, Poland
- Department of Spine Disorders and Pediatric Orthopedics, University of Medical Sciences, Poznan, Poland
| | - Jakub Dalibor Rybka
- Center for Advanced Technology, Adam Mickiewicz University in Poznan, Poznan, Poland.
| |
Collapse
|
38
|
Sharifi MA, Wierer M, Dang TA, Milic J, Moggio A, Sachs N, von Scheidt M, Hinterdobler J, Müller P, Werner J, Stiller B, Aherrahrou Z, Erdmann J, Zaliani A, Graettinger M, Reinshagen J, Gul S, Gribbon P, Maegdefessel L, Bernhagen J, Sager HB, Mann M, Schunkert H, Kessler T. ADAMTS-7 Modulates Atherosclerotic Plaque Formation by Degradation of TIMP-1. Circ Res 2023; 133:674-686. [PMID: 37675562 PMCID: PMC7615141 DOI: 10.1161/circresaha.123.322737] [Citation(s) in RCA: 12] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/06/2023] [Accepted: 08/23/2023] [Indexed: 09/08/2023]
Abstract
BACKGROUND The ADAMTS7 locus was genome-wide significantly associated with coronary artery disease. Lack of the ECM (extracellular matrix) protease ADAMTS-7 (A disintegrin and metalloproteinase-7) was shown to reduce atherosclerotic plaque formation. Here, we sought to identify molecular mechanisms and downstream targets of ADAMTS-7 mediating the risk of atherosclerosis. METHODS Targets of ADAMTS-7 were identified by high-resolution mass spectrometry of atherosclerotic plaques from Apoe-/- and Apoe-/-Adamts7-/- mice. ECM proteins were identified using solubility profiling. Putative targets were validated using immunofluorescence, in vitro degradation assays, coimmunoprecipitation, and Förster resonance energy transfer-based protein-protein interaction assays. ADAMTS7 expression was measured in fibrous caps of human carotid artery plaques. RESULTS In humans, ADAMTS7 expression was higher in caps of unstable as compared to stable carotid plaques. Compared to Apoe-/- mice, atherosclerotic aortas of Apoe-/- mice lacking Adamts-7 (Apoe-/-Adamts7-/-) contained higher protein levels of Timp-1 (tissue inhibitor of metalloprotease-1). In coimmunoprecipitation experiments, the catalytic domain of ADAMTS-7 bound to TIMP-1, which was degraded in the presence of ADAMTS-7 in vitro. ADAMTS-7 reduced the inhibitory capacity of TIMP-1 at its canonical target MMP-9 (matrix metalloprotease-9). As a downstream mechanism, we investigated collagen content in plaques of Apoe-/- and Apoe-/-Adamts7-/- mice after a Western diet. Picrosirius red staining of the aortic root revealed less collagen as a readout of higher MMP-9 activity in Apoe-/- as compared to Apoe-/- Adamts7-/- mice. To facilitate high-throughput screening for ADAMTS-7 inhibitors with the aim of decreasing TIMP-1 degradation, we designed a Förster resonance energy transfer-based assay targeting the ADAMTS-7 catalytic site. CONCLUSIONS ADAMTS-7, which is induced in unstable atherosclerotic plaques, decreases TIMP-1 stability reducing its inhibitory effect on MMP-9, which is known to promote collagen degradation and is likewise associated with coronary artery disease. Disrupting the interaction of ADAMTS-7 and TIMP-1 might be a strategy to increase collagen content and plaque stability for the reduction of atherosclerosis-related events.
Collapse
Affiliation(s)
- M. Amin Sharifi
- Department of Cardiology, German Heart Centre Munich, Technical University of Munich, Munich, Germany
- German Centre for Cardiovascular Research (DZHK e.V.), partner site Munich Heart Alliance, Munich, Germany
| | - Michael Wierer
- Department of Proteomics and Signal Transduction, Max-Planck Institute of Biochemistry, Martinsried, Germany
| | - Tan An Dang
- Department of Cardiology, German Heart Centre Munich, Technical University of Munich, Munich, Germany
- German Centre for Cardiovascular Research (DZHK e.V.), partner site Munich Heart Alliance, Munich, Germany
| | - Jelena Milic
- Division of Vascular Biology, Institute for Stroke and Dementia Research, Ludwig Maximilian University of Munich, Munich, Germany
| | - Aldo Moggio
- Department of Cardiology, German Heart Centre Munich, Technical University of Munich, Munich, Germany
| | - Nadja Sachs
- Vascular Biology and Experimental Vascular Medicine Unit, Department of Vascular and Endovascular Surgery, Klinikum rechts der Isar, Technical University Munich, Munich, Germany
| | - Moritz von Scheidt
- Department of Cardiology, German Heart Centre Munich, Technical University of Munich, Munich, Germany
- German Centre for Cardiovascular Research (DZHK e.V.), partner site Munich Heart Alliance, Munich, Germany
| | - Julia Hinterdobler
- Department of Cardiology, German Heart Centre Munich, Technical University of Munich, Munich, Germany
- German Centre for Cardiovascular Research (DZHK e.V.), partner site Munich Heart Alliance, Munich, Germany
| | - Philipp Müller
- Department of Cardiology, German Heart Centre Munich, Technical University of Munich, Munich, Germany
- German Centre for Cardiovascular Research (DZHK e.V.), partner site Munich Heart Alliance, Munich, Germany
| | - Julia Werner
- Department of Cardiology, German Heart Centre Munich, Technical University of Munich, Munich, Germany
- German Centre for Cardiovascular Research (DZHK e.V.), partner site Munich Heart Alliance, Munich, Germany
| | - Barbara Stiller
- Department of Cardiology, German Heart Centre Munich, Technical University of Munich, Munich, Germany
| | - Zouhair Aherrahrou
- Institute for Cardiogenetics and University Heart Centre Lübeck, University of Lübeck, Lübeck, Germany
- German Centre for Cardiovascular Research (DZHK e.V.), partner site Hamburg/Kiel/Lübeck, Germany
| | - Jeanette Erdmann
- Institute for Cardiogenetics and University Heart Centre Lübeck, University of Lübeck, Lübeck, Germany
- German Centre for Cardiovascular Research (DZHK e.V.), partner site Hamburg/Kiel/Lübeck, Germany
| | - Andrea Zaliani
- Fraunhofer Institute for Translational Medicine and Pharmacology (ITMP), Hamburg, Germany
- Fraunhofer Cluster of Excellence for Immune-Mediated Diseases (CIMD), Hamburg, Germany
| | - Mira Graettinger
- Fraunhofer Institute for Translational Medicine and Pharmacology (ITMP), Hamburg, Germany
- Fraunhofer Cluster of Excellence for Immune-Mediated Diseases (CIMD), Hamburg, Germany
| | - Jeanette Reinshagen
- Fraunhofer Institute for Translational Medicine and Pharmacology (ITMP), Hamburg, Germany
- Fraunhofer Cluster of Excellence for Immune-Mediated Diseases (CIMD), Hamburg, Germany
| | - Sheraz Gul
- Fraunhofer Institute for Translational Medicine and Pharmacology (ITMP), Hamburg, Germany
- Fraunhofer Cluster of Excellence for Immune-Mediated Diseases (CIMD), Hamburg, Germany
| | - Philip Gribbon
- Fraunhofer Institute for Translational Medicine and Pharmacology (ITMP), Hamburg, Germany
- Fraunhofer Cluster of Excellence for Immune-Mediated Diseases (CIMD), Hamburg, Germany
| | - Lars Maegdefessel
- German Centre for Cardiovascular Research (DZHK e.V.), partner site Munich Heart Alliance, Munich, Germany
- Vascular Biology and Experimental Vascular Medicine Unit, Department of Vascular and Endovascular Surgery, Klinikum rechts der Isar, Technical University Munich, Munich, Germany
| | - Jürgen Bernhagen
- German Centre for Cardiovascular Research (DZHK e.V.), partner site Munich Heart Alliance, Munich, Germany
- Division of Vascular Biology, Institute for Stroke and Dementia Research, Ludwig Maximilian University of Munich, Munich, Germany
| | - Hendrik B. Sager
- Department of Cardiology, German Heart Centre Munich, Technical University of Munich, Munich, Germany
- German Centre for Cardiovascular Research (DZHK e.V.), partner site Munich Heart Alliance, Munich, Germany
| | - Matthias Mann
- Department of Proteomics and Signal Transduction, Max-Planck Institute of Biochemistry, Martinsried, Germany
| | - Heribert Schunkert
- Department of Cardiology, German Heart Centre Munich, Technical University of Munich, Munich, Germany
- German Centre for Cardiovascular Research (DZHK e.V.), partner site Munich Heart Alliance, Munich, Germany
| | - Thorsten Kessler
- Department of Cardiology, German Heart Centre Munich, Technical University of Munich, Munich, Germany
- German Centre for Cardiovascular Research (DZHK e.V.), partner site Munich Heart Alliance, Munich, Germany
| |
Collapse
|
39
|
Marciano CL, Hiland TA, Jackson KL, Street S, Maris C, Ehrsam A, Hum JM, Loghmani MT, Chu TMG, Kang KS, Lowery JW. Soft Tissue Manipulation Alters RANTES/CCL5 and IL-4 Cytokine Levels in a Rat Model of Chronic Low Back Pain. Int J Mol Sci 2023; 24:14392. [PMID: 37762698 PMCID: PMC10531608 DOI: 10.3390/ijms241814392] [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: 08/18/2023] [Revised: 09/12/2023] [Accepted: 09/17/2023] [Indexed: 09/29/2023] Open
Abstract
Low back pain (LBP) is a common musculoskeletal complaint that can impede physical function and mobility. Current management often involves pain medication, but there is a need for non-pharmacological and non-invasive interventions. Soft tissue manipulation (STM), such as massage, has been shown to be effective in human subjects, but the molecular mechanisms underlying these findings are not well understood. In this paper, we evaluated potential changes in the soft tissue levels of more than thirty pro- or anti-inflammatory cytokines following instrument-assisted STM (IASTM) in rats with chronic, induced LBP using Complete Freund's Adjuvant. Our results indicate that IASTM is associated with reduced soft tissue levels of Regulated on Activation, Normal T cell Expressed and Secreted (RANTES)/Chemokine (C-C motif) ligand 5 (CCL5) and increased soft tissue levels of Interleukin (IL)-4, which are pro-inflammatory and anti-inflammatory factors, respectively, by 120 min post-treatment. IASTM was not associated with tissue-level changes in C-X-C Motif Chemokine Ligand (CXCL)-5/Lipopolysaccharide-Induced CXC Chemokine (LIX)-which is the murine homologue of IL-8, CXCL-7, Granulocyte-Macrophage-Colony Simulating Factor (GM-CSF), Intercellular Adhesion Molecule (ICAM)-1, IL1-Receptor Antagonist (IL-1ra), IL-6, Interferon-Inducible Protein (IP)-10/CXCL-10, L-selectin, Tumor Necrosis Factor (TNF)-α, or Vascular Endothelial Growth Factor (VEGF) at either 30 or 120 min post-treatment. Combined, our findings raise the possibility that IASTM may exert tissue-level effects associated with improved clinical outcomes and potentially beneficial changes in pro-/anti-inflammatory cytokines in circulation and at the tissue level.
Collapse
Affiliation(s)
- Carmela L. Marciano
- Division of Biomedical Science, College of Osteopathic Medicine, Marian University, Indianapolis, IN 46222, USA; (C.L.M.); (T.A.H.); (S.S.); (A.E.); (J.M.H.)
- Bone & Muscle Research Group, Marian University, Indianapolis, IN 46222, USA; (C.M.); (K.S.K.)
| | - Taylor A. Hiland
- Division of Biomedical Science, College of Osteopathic Medicine, Marian University, Indianapolis, IN 46222, USA; (C.L.M.); (T.A.H.); (S.S.); (A.E.); (J.M.H.)
- Bone & Muscle Research Group, Marian University, Indianapolis, IN 46222, USA; (C.M.); (K.S.K.)
| | - Krista L. Jackson
- Division of Biomedical Science, College of Osteopathic Medicine, Marian University, Indianapolis, IN 46222, USA; (C.L.M.); (T.A.H.); (S.S.); (A.E.); (J.M.H.)
- Bone & Muscle Research Group, Marian University, Indianapolis, IN 46222, USA; (C.M.); (K.S.K.)
| | - Sierra Street
- Division of Biomedical Science, College of Osteopathic Medicine, Marian University, Indianapolis, IN 46222, USA; (C.L.M.); (T.A.H.); (S.S.); (A.E.); (J.M.H.)
- Bone & Muscle Research Group, Marian University, Indianapolis, IN 46222, USA; (C.M.); (K.S.K.)
| | - Carson Maris
- Bone & Muscle Research Group, Marian University, Indianapolis, IN 46222, USA; (C.M.); (K.S.K.)
| | - Andrew Ehrsam
- Division of Biomedical Science, College of Osteopathic Medicine, Marian University, Indianapolis, IN 46222, USA; (C.L.M.); (T.A.H.); (S.S.); (A.E.); (J.M.H.)
- Bone & Muscle Research Group, Marian University, Indianapolis, IN 46222, USA; (C.M.); (K.S.K.)
| | - Julia M. Hum
- Division of Biomedical Science, College of Osteopathic Medicine, Marian University, Indianapolis, IN 46222, USA; (C.L.M.); (T.A.H.); (S.S.); (A.E.); (J.M.H.)
- Bone & Muscle Research Group, Marian University, Indianapolis, IN 46222, USA; (C.M.); (K.S.K.)
- Indiana Biosciences Research Institute, Indianapolis, IN 46222, USA
| | - Mary Terry Loghmani
- Department of Physical Therapy, School of Health and Human Sciences, Indiana University, Indianapolis, IN 46222, USA;
- Indiana Center for Musculoskeletal Health, School of Medicine, Indiana University, Indianapolis, IN 46222, USA;
| | - Tien-Min G. Chu
- Indiana Center for Musculoskeletal Health, School of Medicine, Indiana University, Indianapolis, IN 46222, USA;
- Department of Biomedical Sciences and Comprehensive Care, School of Dentistry, Indiana University, Indianapolis, IN 46222, USA
| | - Kyung S. Kang
- Bone & Muscle Research Group, Marian University, Indianapolis, IN 46222, USA; (C.M.); (K.S.K.)
- Indiana Biosciences Research Institute, Indianapolis, IN 46222, USA
- Witchger School of Engineering, Marian University, Indianapolis, IN 46222, USA
| | - Jonathan W. Lowery
- Division of Biomedical Science, College of Osteopathic Medicine, Marian University, Indianapolis, IN 46222, USA; (C.L.M.); (T.A.H.); (S.S.); (A.E.); (J.M.H.)
- Bone & Muscle Research Group, Marian University, Indianapolis, IN 46222, USA; (C.M.); (K.S.K.)
- Indiana Biosciences Research Institute, Indianapolis, IN 46222, USA
- Indiana Center for Musculoskeletal Health, School of Medicine, Indiana University, Indianapolis, IN 46222, USA;
- Division of Academic Affairs, Marian University, Indianapolis, IN 46222, USA
- Department of Orthopaedic Surgery, School of Medicine, Indiana University, Indianapolis, IN 46222, USA
| |
Collapse
|
40
|
Wang X, Wang M, Zhou Z, Zou X, Song G, Zhang Q, Zhou H. SMOC2 promoted vascular smooth muscle cell proliferation, migration, and extracellular matrix degradation by activating BMP/TGF-β1 signaling pathway. J Clin Biochem Nutr 2023; 73:116-123. [PMID: 37700850 PMCID: PMC10493216 DOI: 10.3164/jcbn.22-100] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2022] [Accepted: 01/04/2023] [Indexed: 09/14/2023] Open
Abstract
A widespread degenerative condition of the aorta, abdominal aortic aneurysm (AAA), severely endangers the health of middle-aged and elderly people. SPARC related modular calcium binding2 (SMOC2) is upregulated in the carotid arteries of rats with atherosclerotic lesions, but its function in AAA is still unknown. Therefore, the aim of this research was to evaluate the function of SMOC2 in AAA. The results showed that in the AAA tissues, SMOC2 expression was upregulated compared with healthy controls. Overexpression of SMOC2 promoted vascular smooth muscle cells (VSMCs) proliferation, migration, and extracellular matrix (ECM) degradation. In contrast, silence of SMOC2 inhibited VSMCs proliferation, migration, and ECM degradation. Overexpression of SMOC2 promoted BMP and TGF-β1 expression and silence of SMOC2 had an opposite effect. Besides, inhibition of BMP or TGF-β1 suppressed VSMCs cell proliferation, migration, and ECM degradation. Moreover, inhibition BMP or TGF-β1 reversed the promotive effects of SMOC2 overexpression on VSMCs proliferation, migration, and ECM degradation. SMOC2 may affecte the formation of AAA by upregulating BMP and TGF-β1 to regulate the proliferation, migration, and ECM degradation of VSMCs.
Collapse
Affiliation(s)
- Xiaowei Wang
- Department of Vascular Surgery, Weihai Municipal Hospital, Cheeloo College of Medicine, Shandong University, No. 70 Heping Road, Huancui District, Weihai, Shandong 264200, China
| | - Meng Wang
- Department of Nephrology, Weihai Municipal Hospital, Cheeloo College of Medicine, Shandong University, No. 70 Heping Road, Huancui District, Weihai, Shandong 264200, China
| | - Zhongxiao Zhou
- Department of Vascular Surgery, Weihai Municipal Hospital, Cheeloo College of Medicine, Shandong University, No. 70 Heping Road, Huancui District, Weihai, Shandong 264200, China
| | - Xin Zou
- Department of Vascular Surgery, Weihai Municipal Hospital, Cheeloo College of Medicine, Shandong University, No. 70 Heping Road, Huancui District, Weihai, Shandong 264200, China
| | - Guoxin Song
- Department of Vascular Surgery, Weihai Municipal Hospital, Cheeloo College of Medicine, Shandong University, No. 70 Heping Road, Huancui District, Weihai, Shandong 264200, China
| | - Qingsong Zhang
- Department of Vascular Surgery, Weihai Municipal Hospital, Cheeloo College of Medicine, Shandong University, No. 70 Heping Road, Huancui District, Weihai, Shandong 264200, China
| | - Haimeng Zhou
- Department of Vascular Surgery, Weihai Municipal Hospital, Cheeloo College of Medicine, Shandong University, No. 70 Heping Road, Huancui District, Weihai, Shandong 264200, China
| |
Collapse
|
41
|
Mubtasim N, Gollahon L. Characterizing 3T3-L1 MBX Adipocyte Cell Differentiation Maintained with Fatty Acids as an In Vitro Model to Study the Effects of Obesity. Life (Basel) 2023; 13:1712. [PMID: 37629569 PMCID: PMC10455818 DOI: 10.3390/life13081712] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2023] [Revised: 07/31/2023] [Accepted: 08/03/2023] [Indexed: 08/27/2023] Open
Abstract
The increasing prevalence of obesity has prompted intensive research into understanding its role in pathogenesis and designing appropriate treatments. To determine the signals generated from the interaction of fat cells with a target organ, a reliable white adipocyte model in vitro is needed. Differentiated fibroblasts are the most extensively studied using in vitro cell models of white adipocytes. However, it can be argued that differentiated fibroblasts minimally recapitulate the consequences of obesity. Here, we describe 3T3-L1 MBX cells as a culture model for studying obese adipocytes and their effects. Differentiation of 3T3-L1 MBX cells was at first optimized and then maintained in the presence of fatty acids cocktail combination to induce the obese condition. Lipid accumulation and adipokine secretion profiles were analyzed. Results showed that fatty acid-maintained, differentiated 3T3-L1 MBX cells had significantly greater accumulation of lipids and significant changes in the adipokine secretions in comparison to differentiated 3T3-L1 MBX cells maintained in medium without fatty acids. To elucidate the molecular changes associated with adipogenesis and lipid accumulation profile of 3T3-L1 MBX cells, we have also explored the expression of some of the regulatory proteins related to the development and maintenance of adipocytes from the preadipocyte lineage.
Collapse
Affiliation(s)
| | - Lauren Gollahon
- Department of Biological Sciences, Texas Tech University, 2500 Broadway, Lubbock, TX 79409, USA;
| |
Collapse
|
42
|
Choi JK, Chung H, Oh SJ, Kim JW, Kim SH. Functionally enhanced cell spheroids for stem cell therapy: Role of TIMP1 in the survival and therapeutic effectiveness of stem cell spheroids. Acta Biomater 2023; 166:454-469. [PMID: 37230438 DOI: 10.1016/j.actbio.2023.05.033] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2023] [Revised: 05/16/2023] [Accepted: 05/18/2023] [Indexed: 05/27/2023]
Abstract
Stem cell therapy has emerged as a promising regenerative medicine strategy but is limited by poor cell survival, leading to low therapeutic outcomes. We developed cell spheroid therapeutics to overcome this limitation. We utilized solid-phase FGF2 to form functionally enhanced cell spheroid-adipose derived (FECS-Ad), a type of cell spheroid that preconditions cells with intrinsic hypoxia to increase the survival of transplanted cells. We demonstrated an increase in hypoxia-inducible factor 1-alpha (HIF-1α) levels in FECS-Ad, which led to the upregulation of tissue inhibitor of metalloproteinase 1 (TIMP1). TIMP1 enhanced the survival of FECS-Ad, presumably through the CD63/FAK/Akt/Bcl2 anti-apoptotic signaling pathway. Cell viability of transplanted FECS-Ad was reduced by TIMP1 knockdown in an in vitro collagen gel block and a mouse model of critical limb ischemia (CLI). TIMP1 knockdown in FECS-Ad inhibited angiogenesis and muscle regeneration induced by FECS-Ad transplanted into ischemic mouse tissue. Genetic overexpression of TIMP1 in FECS-Ad further promoted the survival and therapeutic efficacy of transplanted FECS-Ad. Collectively, we suggest that TIMP1 acts as a key survival factor to improve the survival of transplanted stem cell spheroids, which provides scientific evidence for enhanced therapeutic efficacy of stem cell spheroids, and FECS-Ad as a potential therapeutic agent to treat CLI. STATEMENT OF SIGNIFICANCE: We used FGF2-tethered substrate platform to form adipose-derived stem cell spheroids, as we named as functionally enhanced cell spheroid-adipose derived (FECS-Ad). In this paper, we showed that intrinsic hypoxia of spheroids upregulated expression of HIF-1α, which in turn upregulated expression of TIMP1. Our paper highlights TIMP1 as a key survival factor to improve survival of transplanted stem cell spheroids. We believe that our study has a very strong scientific impact as extending transplantation efficiency is essential for successful stem cell therapy.
Collapse
Affiliation(s)
- Jung-Kyun Choi
- Center for Biomaterials, Biomedical Research Institute, Korea Institute of Science and Technology (KIST), Seoul 02792, Republic of Korea; Division of Bio-Medical Science & Technology, KIST School, University of Science and Technology, Seoul 02792, Republic of Korea
| | - Haeun Chung
- Center for Biomaterials, Biomedical Research Institute, Korea Institute of Science and Technology (KIST), Seoul 02792, Republic of Korea; Division of Bio-Medical Science & Technology, KIST School, University of Science and Technology, Seoul 02792, Republic of Korea
| | - Seung Ja Oh
- Department of Genetics and Biotechnology, College of Life Sciences, Kyung Hee University, Yongin-si, Gyeonggi-do, 17104, Republic of Korea
| | - Jong-Wan Kim
- S.Biomedics Co., Ltd., Seoul 04797, Republic of Korea
| | - Sang-Heon Kim
- Center for Biomaterials, Biomedical Research Institute, Korea Institute of Science and Technology (KIST), Seoul 02792, Republic of Korea; Division of Bio-Medical Science & Technology, KIST School, University of Science and Technology, Seoul 02792, Republic of Korea.
| |
Collapse
|
43
|
Ebert S, Zang L, Ismail N, Otabil M, Fröhlich A, Egea V, Ács S, Hoeberg M, Berres ML, Weber C, Moreira JMA, Ries C, Bernhagen J, El Bounkari O. Tissue Inhibitor of Metalloproteinases-1 Interacts with CD74 to Promote AKT Signaling, Monocyte Recruitment Responses, and Vascular Smooth Muscle Cell Proliferation. Cells 2023; 12:1899. [PMID: 37508563 PMCID: PMC10378328 DOI: 10.3390/cells12141899] [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: 06/13/2023] [Revised: 07/14/2023] [Accepted: 07/17/2023] [Indexed: 07/30/2023] Open
Abstract
Tissue inhibitor of metalloproteinases-1 (TIMP-1), an important regulator of matrix metalloproteinases (MMPs), has recently been shown to interact with CD74, a receptor for macrophage migration inhibitory factor (MIF). However, the biological effects mediated by TIMP-1 through CD74 remain largely unexplored. Using sequence alignment and in silico protein-protein docking analysis, we demonstrated that TIMP-1 shares residues with both MIF and MIF-2, crucial for CD74 binding, but not for CXCR4. Subcellular colocalization, immunoprecipitation, and internalization experiments supported these findings, demonstrating that TIMP-1 interacts with surface-expressed CD74, resulting in its internalization in a dose-dependent manner, as well as with a soluble CD74 ectodomain fragment (sCD74). This prompted us to study the effects of the TIMP-1-CD74 axis on monocytes and vascular smooth muscle cells (VSCMs) to assess its impact on vascular inflammation. A phospho-kinase array revealed the activation of serine/threonine kinases by TIMP-1 in THP-1 pre-monocytes, in particular AKT. Similarly, TIMP-1 dose-dependently triggered the phosphorylation of AKT and ERK1/2 in primary human monocytes. Importantly, Transwell migration, 3D-based Chemotaxis, and flow adhesion assays demonstrated that TIMP-1 engagement of CD74 strongly promotes the recruitment response of primary human monocytes, while live cell imaging studies revealed a profound activating effect on VSMC proliferation. Finally, re-analysis of scRNA-seq data highlighted the expression patterns of TIMP-1 and CD74 in human atherosclerotic lesions, thus, together with our experimental data, indicating a role for the TIMP-1-CD74 axis in vascular inflammation and atherosclerosis.
Collapse
Affiliation(s)
- Simon Ebert
- Department of Vascular Biology, Institute for Stroke and Dementia Research, Klinikum der Universität München, Ludwig-Maximilian-University (LMU) Munich, 81377 Munich, Germany
| | - Lan Zang
- Institute for Cardiovascular Prevention (IPEK), Klinikum der Universität München, Ludwig-Maximilian-University (LMU) Munich, 80336 Munich, Germany
| | - Noor Ismail
- Department of Vascular Biology, Institute for Stroke and Dementia Research, Klinikum der Universität München, Ludwig-Maximilian-University (LMU) Munich, 81377 Munich, Germany
| | - Michael Otabil
- Department of Vascular Biology, Institute for Stroke and Dementia Research, Klinikum der Universität München, Ludwig-Maximilian-University (LMU) Munich, 81377 Munich, Germany
| | - Adrian Fröhlich
- Department of Vascular Biology, Institute for Stroke and Dementia Research, Klinikum der Universität München, Ludwig-Maximilian-University (LMU) Munich, 81377 Munich, Germany
| | - Virginia Egea
- Institute for Cardiovascular Prevention (IPEK), Klinikum der Universität München, Ludwig-Maximilian-University (LMU) Munich, 80336 Munich, Germany
| | - Susann Ács
- Institute for Cardiovascular Prevention (IPEK), Klinikum der Universität München, Ludwig-Maximilian-University (LMU) Munich, 80336 Munich, Germany
| | - Mikkel Hoeberg
- Department of Drug Design and Pharmacology, Faculty of Health and Medical Sciences, University of Copenhagen, 2200 Copenhagen, Denmark
| | - Marie-Luise Berres
- Department of Internal Medicine III, RWTH Aachen University, 52074 Aachen, Germany
| | - Christian Weber
- Institute for Cardiovascular Prevention (IPEK), Klinikum der Universität München, Ludwig-Maximilian-University (LMU) Munich, 80336 Munich, Germany
- Munich Cluster for Systems Neurology (SyNergy), 81377 Munich, Germany
- Munich Heart Alliance, 80802 Munich, Germany
| | - José M A Moreira
- Department of Drug Design and Pharmacology, Faculty of Health and Medical Sciences, University of Copenhagen, 2200 Copenhagen, Denmark
| | - Christian Ries
- Institute for Cardiovascular Prevention (IPEK), Klinikum der Universität München, Ludwig-Maximilian-University (LMU) Munich, 80336 Munich, Germany
| | - Jürgen Bernhagen
- Department of Vascular Biology, Institute for Stroke and Dementia Research, Klinikum der Universität München, Ludwig-Maximilian-University (LMU) Munich, 81377 Munich, Germany
- Munich Cluster for Systems Neurology (SyNergy), 81377 Munich, Germany
- Munich Heart Alliance, 80802 Munich, Germany
| | - Omar El Bounkari
- Department of Vascular Biology, Institute for Stroke and Dementia Research, Klinikum der Universität München, Ludwig-Maximilian-University (LMU) Munich, 81377 Munich, Germany
| |
Collapse
|
44
|
Liu T, Bai M, Liu M, Li T, Liao Y, Zhao C, Yao M, Wang J, Wen A, Ding Y. Novel synergistic mechanism of 11-keto-β-boswellic acid and Z-Guggulsterone on ischemic stroke revealed by single-cell transcriptomics. Pharmacol Res 2023; 193:106803. [PMID: 37230158 DOI: 10.1016/j.phrs.2023.106803] [Citation(s) in RCA: 21] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/26/2023] [Revised: 05/11/2023] [Accepted: 05/22/2023] [Indexed: 05/27/2023]
Abstract
Although strides have been made, the challenge of preventing and treating ischemic stroke continues to persist globally. For thousands of years, the natural substances Frankincense and Myrrh have been employed in Chinese and Indian medicine to address cerebrovascular diseases, with the key components of 11-keto-β-boswellic acid (KBA) and Z-Guggulsterone (Z-GS) being the active agents. In this study, the synergistic effect and underlying mechanism of KBA and Z-GS on ischemic stroke were examined using single-cell transcriptomics. Fourteen cell types were identified in KBA-Z-GS-treated ischemic penumbra, and microglia and astrocytes account for the largest proportion. They were further re-clustered into six and seven subtypes, respectively. GSVA analysis reflected the distinct roles of each subtype. Pseudo-time trajectory indicated that Slc1a2 and Timp1 were core fate transition genes regulated by KBA-Z-GS. In addition, KBA-Z-GS synergistically regulated inflammatory reactions in microglia and cellular metabolism and ferroptosis in astrocytes. Most notably, we established an innovative drug-gene synergistic regulation pattern, and genes regulated by KBA-Z-GS were divided into four categories based on this pattern. Finally, Spp1 was demonstrated as the hub target of KBA-Z-GS. Taken together, this study reveals the synergistic mechanism of KBA and Z-GS on cerebral ischemia, and Spp1 may be the synergistic target for that. Precise drug development targeting Spp1 may offer a potential therapeutic approach for treating ischemic stroke.
Collapse
Affiliation(s)
- Tianlong Liu
- Department of Pharmacy, Xijing Hospital, Fourth Military Medical University, Xi'an 710032, China; Department of Pharmacy, The 940th Hospital Joint Logistics Support Forces of PLA, Lanzhou 730050, China
| | - Min Bai
- Department of Pharmacy, Xijing Hospital, Fourth Military Medical University, Xi'an 710032, China
| | - Minna Liu
- Department of Nephrology, The 940th Hospital Joint Logistics Support Forces of PLA, Lanzhou 730050, China
| | - Tian Li
- School of Basic Medicine, Fourth Military Medical University, Xi'an 710032, China
| | - Yucheng Liao
- Department of Pharmacy, Xijing Hospital, Fourth Military Medical University, Xi'an 710032, China
| | - Chao Zhao
- Department of Pharmacy, Xijing Hospital, Fourth Military Medical University, Xi'an 710032, China
| | - Minna Yao
- Department of Pharmacy, Xijing Hospital, Fourth Military Medical University, Xi'an 710032, China
| | - Jingwen Wang
- Department of Pharmacy, Xijing Hospital, Fourth Military Medical University, Xi'an 710032, China.
| | - Aidong Wen
- Department of Pharmacy, Xijing Hospital, Fourth Military Medical University, Xi'an 710032, China.
| | - Yi Ding
- Department of Pharmacy, Xijing Hospital, Fourth Military Medical University, Xi'an 710032, China.
| |
Collapse
|
45
|
Peeney D, Fan Y, Gurung S, Lazaroff C, Ratnayake S, Warner A, Karim B, Meerzaman D, Stetler-Stevenson WG. Whole organism profiling of the Timp gene family. Matrix Biol Plus 2023; 18:100132. [PMID: 37095886 PMCID: PMC10121480 DOI: 10.1016/j.mbplus.2023.100132] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2023] [Revised: 03/28/2023] [Accepted: 03/31/2023] [Indexed: 04/04/2023] Open
Abstract
Tissue inhibitor of metalloproteinases (TIMPs/Timps) are an endogenous family of widely expressed matrisome-associated proteins that were initially identified as inhibitors of matrix metalloproteinase activity (Metzincin family proteases). Consequently, TIMPs are often considered simply as protease inhibitors by many investigators. However, an evolving list of new metalloproteinase-independent functions for TIMP family members suggests that this concept is outdated. These novel TIMP functions include direct agonism/antagonism of multiple transmembrane receptors, as well as functional interactions with matrisome targets. While the family was fully identified over two decades ago, there has yet to be an in-depth study describing the expression of TIMPs in normal tissues of adult mammals. An understanding of the tissues and cell-types that express TIMPs 1 through 4, in both normal and disease states are important to contextualize the growing functional capabilities of TIMP proteins, which are often dismissed as non-canonical. Using publicly available single cell RNA sequencing data from the Tabula Muris Consortium, we analyzed approximately 100,000 murine cells across eighteen tissues from non-diseased organs, representing seventy-three annotated cell types, to define the diversity in Timp gene expression across healthy tissues. We describe the unique expression profiles across tissues and organ-specific cell types that all four Timp genes display. Within annotated cell-types, we identify clear and discrete cluster-specific patterns of Timp expression, particularly in cells of stromal and endothelial origins. RNA in-situ hybridization across four organs expands on the scRNA sequencing analysis, revealing novel compartments associated with individual Timp expression. These analyses emphasize a need for specific studies investigating the functional significance of Timp expression in the identified tissues and cell sub-types. This understanding of the tissues, specific cell types and microenvironment conditions in which Timp genes are expressed adds important physiological context to the growing array of novel functions for TIMP proteins.
Collapse
Affiliation(s)
- David Peeney
- Extracellular Matrix Pathology Section, Laboratory of Pathology, National Cancer Institute, National Institute of Health, Bethesda, MD, USA
| | - Yu Fan
- Computational Genomics and Bioinformatics Branch, Center for Biomedical Informatics & Information Technology, National Cancer Institute, National Institute of Health, Rockville, MD, USA
| | - Sadeechya Gurung
- Extracellular Matrix Pathology Section, Laboratory of Pathology, National Cancer Institute, National Institute of Health, Bethesda, MD, USA
| | - Carolyn Lazaroff
- Extracellular Matrix Pathology Section, Laboratory of Pathology, National Cancer Institute, National Institute of Health, Bethesda, MD, USA
| | - Shashikala Ratnayake
- Computational Genomics and Bioinformatics Branch, Center for Biomedical Informatics & Information Technology, National Cancer Institute, National Institute of Health, Rockville, MD, USA
| | - Andrew Warner
- Molecular Histopathology Laboratory, Frederick National Laboratory, National Cancer Institute, Frederick, MD, USA
| | - Baktiar Karim
- Molecular Histopathology Laboratory, Frederick National Laboratory, National Cancer Institute, Frederick, MD, USA
| | - Daoud Meerzaman
- Computational Genomics and Bioinformatics Branch, Center for Biomedical Informatics & Information Technology, National Cancer Institute, National Institute of Health, Rockville, MD, USA
| | - William G. Stetler-Stevenson
- Extracellular Matrix Pathology Section, Laboratory of Pathology, National Cancer Institute, National Institute of Health, Bethesda, MD, USA
| |
Collapse
|
46
|
Wright K, Ly T, Kriet M, Czirok A, Thomas SM. Cancer-Associated Fibroblasts: Master Tumor Microenvironment Modifiers. Cancers (Basel) 2023; 15:cancers15061899. [PMID: 36980785 PMCID: PMC10047485 DOI: 10.3390/cancers15061899] [Citation(s) in RCA: 56] [Impact Index Per Article: 28.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2023] [Revised: 03/17/2023] [Accepted: 03/20/2023] [Indexed: 03/30/2023] Open
Abstract
Cancer cells rely on the tumor microenvironment (TME), a composite of non-malignant cells, and extracellular matrix (ECM), for survival, growth, and metastasis. The ECM contributes to the biomechanical properties of the surrounding tissue, in addition to providing signals for tissue development. Cancer-associated fibroblasts (CAFs) are stromal cells in the TME that are integral to cancer progression. Subtypes of CAFs across a variety of cancers have been revealed, and each play a different role in cancer progression or suppression. CAFs secrete signaling molecules and remodel the surrounding ECM by depositing its constituents as well as degrading enzymes. In cancer, a remodeled ECM can lead to tumor-promoting effects. Not only does the remodeled ECM promote growth and allow for easier metastasis, but it can also modulate the immune system. A better understanding of how CAFs remodel the ECM will likely yield novel therapeutic targets. In this review, we summarize the key factors secreted by CAFs that facilitate tumor progression, ECM remodeling, and immune suppression.
Collapse
Affiliation(s)
- Kellen Wright
- Department of Otolaryngology, University of Kansas Medical Center, Kansas City, KS 66160, USA
| | - Thuc Ly
- Department of Cancer Biology, University of Kansas Medical Center, Kansas City, KS 66160, USA
| | - Matthew Kriet
- Department of Otolaryngology, University of Kansas Medical Center, Kansas City, KS 66160, USA
| | - Andras Czirok
- Department of Cell Biology and Physiology, University of Kansas Medical Center, Kansas City, KS 66160, USA
| | - Sufi Mary Thomas
- Department of Otolaryngology, University of Kansas Medical Center, Kansas City, KS 66160, USA
- Department of Cancer Biology, University of Kansas Medical Center, Kansas City, KS 66160, USA
- Department of Cell Biology and Physiology, University of Kansas Medical Center, Kansas City, KS 66160, USA
| |
Collapse
|
47
|
Egea V, Megens RTA, Santovito D, Wantha S, Brandl R, Siess W, Khani S, Soehnlein O, Bartelt A, Weber C, Ries C. Properties and fate of human mesenchymal stem cells upon miRNA let-7f-promoted recruitment to atherosclerotic plaques. Cardiovasc Res 2023; 119:155-166. [PMID: 35238350 PMCID: PMC10022860 DOI: 10.1093/cvr/cvac022] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/30/2021] [Accepted: 02/28/2022] [Indexed: 11/12/2022] Open
Abstract
AIMS Atherosclerosis is a chronic inflammatory disease of the arteries leading to the formation of atheromatous plaques. Human mesenchymal stem cells (hMSCs) are recruited from the circulation into plaques where in response to their environment they adopt a phenotype with immunomodulatory properties. However, the mechanisms underlying hMSC function in these processes are unclear. Recently, we described that miRNA let-7f controls hMSC invasion guided by inflammatory cytokines and chemokines. Here, we investigated the role of let-7f in hMSC tropism to human atheromas and the effects of the plaque microenvironment on cell fate and release of soluble factors. METHODS AND RESULTS Incubation of hMSCs with LL-37, an antimicrobial peptide abundantly found in plaques, increased biosynthesis of let-7f and N-formyl peptide receptor 2 (FPR2), enabling chemotactic invasion of the cells towards LL-37, as determined by qRT-PCR, flow cytometry, and cell invasion assay analysis. In an Apoe-/- mouse model of atherosclerosis, circulating hMSCs preferentially adhered to athero-prone endothelium. This property was facilitated by elevated levels of let-7f in the hMSCs, as assayed by ex vivo artery perfusion and two-photon laser scanning microscopy. Exposure of hMSCs to homogenized human atheromatous plaque material considerably induced the production of various cytokines, chemokines, matrix metalloproteinases, and tissue inhibitors of metalloproteinases, as studied by PCR array and western blot analysis. Moreover, exposure to human plaque extracts elicited differentiation of hMSCs into cells of the myogenic lineage, suggesting a potentially plaque-stabilizing effect. CONCLUSIONS Our findings indicate that let-7f promotes hMSC tropism towards atheromas through the LL-37/FPR2 axis and demonstrate that hMSCs upon contact with human plaque environment develop a potentially athero-protective signature impacting the pathophysiology of atherosclerosis.
Collapse
Affiliation(s)
- Virginia Egea
- Corresponding authors. Tel: +49-89-4400-55310, E-mail: (C.R.); Tel: +49-89-4400-43902, E-mail: (V.E.)
| | - Remco Theodorus Adrianus Megens
- Institute for Cardiovascular Prevention (IPEK), Ludwig-Maximilians-University of Munich, Munich, Germany
- German Center for Cardiovascular Research (DZHK), Partner Site Munich Heart Alliance, Munich, Germany
- Department of Biomedical Engineering, Cardiovascular Research Institute Maastricht, University of Maastricht, Maastricht, The Netherlands
| | - Donato Santovito
- Institute for Cardiovascular Prevention (IPEK), Ludwig-Maximilians-University of Munich, Munich, Germany
- German Center for Cardiovascular Research (DZHK), Partner Site Munich Heart Alliance, Munich, Germany
- Institute for Genetic and Biomedical Research (IRGB), UoS of Milan, National Research Council (CNR), Milan, Italy
| | - Sarawuth Wantha
- Institute for Cardiovascular Prevention (IPEK), Ludwig-Maximilians-University of Munich, Munich, Germany
| | - Richard Brandl
- St. Mary’s Square Institute for Vascular Surgery and Phlebology, Munich, Germany
| | - Wolfgang Siess
- Institute for Cardiovascular Prevention (IPEK), Ludwig-Maximilians-University of Munich, Munich, Germany
| | - Sajjad Khani
- Institute for Cardiovascular Prevention (IPEK), Ludwig-Maximilians-University of Munich, Munich, Germany
| | - Oliver Soehnlein
- Institute for Cardiovascular Prevention (IPEK), Ludwig-Maximilians-University of Munich, Munich, Germany
- Department of Physiology and Pharmacology (FyFa), Karolinska Institutet, Stockholm, Sweden
- Institute for Experimental Pathology (ExPat), Center for Molecular Biology of Inflammation (ZMBE), Westfaelische Wilhelms-University of Muenster, Muenster, Germany
| | - Alexander Bartelt
- Institute for Cardiovascular Prevention (IPEK), Ludwig-Maximilians-University of Munich, Munich, Germany
- German Center for Cardiovascular Research (DZHK), Partner Site Munich Heart Alliance, Munich, Germany
- Institute for Diabetes and Cancer (IDC), Helmholtz Center Munich, Neuherberg, Germany
- Department of Molecular Metabolism, Sabri Ülker Center for Metabolic Research, Harvard T.H. Chan School of Public Health, 665 Huntington Avenue, Boston, MA 02115, USA
| | - Christian Weber
- Institute for Cardiovascular Prevention (IPEK), Ludwig-Maximilians-University of Munich, Munich, Germany
- Department of Biochemistry, Cardiovascular Research Institute Maastricht, University of Maastricht, Maastricht, The Netherlands
- Munich Cluster for Systems Neurology (SyNergy), Munich, Germany
| | - Christian Ries
- Corresponding authors. Tel: +49-89-4400-55310, E-mail: (C.R.); Tel: +49-89-4400-43902, E-mail: (V.E.)
| |
Collapse
|
48
|
Du T, Yu X, Shao S, Li T, Xu S, Wu L. Aging of Nanoplastics Significantly Affects Protein Corona Composition Thus Enhancing Macrophage Uptake. ENVIRONMENTAL SCIENCE & TECHNOLOGY 2023; 57:3206-3217. [PMID: 36730723 DOI: 10.1021/acs.est.2c05772] [Citation(s) in RCA: 29] [Impact Index Per Article: 14.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/18/2023]
Abstract
Nanoplastics (NPs), as emerging contaminants, have attracted increasing attention for their effects on human exposure and potential health risks. The protein corona formed on the surface of NPs affects the biological activity and fate of the NPs in vivo. However, how environmental aging, an inevitable process once NPs enter the environment, affects the formation of protein corona on NPs is still unclear. This study investigated the changes in the compositions of protein corona formed on photo-aged polystyrene (PS) NPs in human bronchoalveolar lavage fluid (BALF), corresponding to the inhalation exposure pathway. The results demonstrated that both the species and abundance of proteins in the BALF protein corona on the surface of PS NPs were altered by aging. In addition, the aged PS NPs are more hydrophilic and less electronegative than the pristine PS NPs; hence, there is an increased sorption of more negatively charged hydrophilic proteins. Moreover, aging-induced alterations in BALF protein corona enhanced the uptake of aged PS NPs by lung macrophages J774A.1 through phagocytosis and clathrin-mediated endocytosis. These findings highlight the importance of environmental aging processes in the biosafety assessment of nanoplastics.
Collapse
Affiliation(s)
- Tingting Du
- Information Materials and Intelligent Sensing Laboratory of Anhui Province, Institutes of Physical Science and Information Technology, Anhui University, Hefei 230601, China
| | - Xiang Yu
- Information Materials and Intelligent Sensing Laboratory of Anhui Province, Institutes of Physical Science and Information Technology, Anhui University, Hefei 230601, China
| | - Song Shao
- Information Materials and Intelligent Sensing Laboratory of Anhui Province, Institutes of Physical Science and Information Technology, Anhui University, Hefei 230601, China
| | - Tong Li
- Information Materials and Intelligent Sensing Laboratory of Anhui Province, Institutes of Physical Science and Information Technology, Anhui University, Hefei 230601, China
| | - Shengmin Xu
- Information Materials and Intelligent Sensing Laboratory of Anhui Province, Institutes of Physical Science and Information Technology, Anhui University, Hefei 230601, China
| | - Lijun Wu
- Information Materials and Intelligent Sensing Laboratory of Anhui Province, Institutes of Physical Science and Information Technology, Anhui University, Hefei 230601, China
| |
Collapse
|
49
|
Cell-Free Therapies: The Use of Cell Extracts to Mitigate Irradiation-Injured Salivary Glands. BIOLOGY 2023; 12:biology12020305. [PMID: 36829582 PMCID: PMC9953449 DOI: 10.3390/biology12020305] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 01/11/2023] [Revised: 02/06/2023] [Accepted: 02/07/2023] [Indexed: 02/17/2023]
Abstract
Radiotherapy is a standard treatment for head and neck cancer patients worldwide. However, millions of patients who received radiotherapy consequently suffer from xerostomia because of irreversible damage to salivary glands (SGs) caused by irradiation (IR). Current treatments for IR-induced SG hypofunction only provide temporary symptom alleviation but do not repair the damaged SG, thus resulting in limited treatment efficacy. Therefore, there has recently been a growing interest in regenerative treatments, such as cell-free therapies. This review aims to summarize cell-free therapies for IR-induced SG, with a particular emphasis on utilizing diverse cell extract (CE) administrations. Cell extract is a group of heterogeneous mixtures containing multifunctional inter-cellular molecules. This review discusses the current knowledge of CE's components and efficacy. We propose optimal approaches to improve cell extract treatment from multiple perspectives (e.g., delivery routes, preparation methods, and other details regarding CE administration). In addition, the advantages and limitations of CE treatment are systematically discussed by comparing it to other cell-free (such as conditioned media and exosomes) and cell-based therapies. Although a comprehensive identification of the bioactive factors within CEs and their mechanisms of action have yet to be fully understood, we propose cell extract therapy as an effective, practical, user-friendly, and safe option to conventional therapies in IR-induced SG.
Collapse
|
50
|
Shu J, Shi J, Gu Y, Deng L, Zhao C, Wu C, Zhao J, Wang H, Jin L. Levocarnitine regulates the growth of angiotensin II-induced myocardial fibrosis cells via TIMP-1. Open Life Sci 2023; 18:20220554. [PMID: 36816804 PMCID: PMC9922061 DOI: 10.1515/biol-2022-0554] [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: 06/20/2022] [Revised: 11/03/2022] [Accepted: 11/30/2022] [Indexed: 02/11/2023] Open
Abstract
This study aimed to explore the effects of tissue inhibitor of metalloproteinases-1 (TIMP-1) on levocarnitine (LC)-mediated regulation of angiotensin II (AngII)-induced myocardial fibrosis (MF) and its underlying mechanisms. H9C2 cells were treated with AngII for 24 h to induce fibrosis. The cells were then treated with LC or transfected with TIMP-1-OE plasmid/si‑TIMP-1. Cell apoptosis, viability, migration, and related gene expression were analyzed. AngII treatment significantly upregulated Axl, α-SMA, and MMP3 expression (P < 0.05) and downregulated STAT4 and TIMP1 expression (P < 0.05) relative to the control levels. After transfection, cells with TIMP-1 overexpression/knockdown were successfully established. Compared with that of the control, AngII significantly inhibited cell viability and cell migration while promoting cell apoptosis (P < 0.05). LC and TIMP-1-OE transfection further suppressed cell viability and migration induced by Ang II and upregulated apoptosis, whereas si-TIMP-1 had the opposite effect. Furthermore, LC and TIMP-1-OE transfection downregulated Axl, AT1R, α-SMA, collagen III, Bcl-2, and MMP3 expression caused by AngII and upregulated caspase 3, p53, and STAT4 expression, whereas si-TIMP-1 had the opposite effect. TIMP-1 is therefore a potential therapeutic target for delaying MF progression.
Collapse
Affiliation(s)
- Jin Shu
- Department of Gerontology, Shibei Hospital of Jing’an District, Shanghai, 200443, China
| | - Jue Shi
- Department of Gerontology, Shibei Hospital of Jing’an District, Shanghai, 200443, China
| | - Yiwen Gu
- Department of Gerontology, Shibei Hospital of Jing’an District, Shanghai, 200443, China
| | - Lei Deng
- Department of Gerontology, Shibei Hospital of Jing’an District, Shanghai, 200443, China
| | - Chen Zhao
- Department of Gerontology, Shibei Hospital of Jing’an District, Shanghai, 200443, China
| | - Chun Wu
- Department of Gerontology, Shibei Hospital of Jing’an District, Shanghai, 200443, China
| | - Jiachen Zhao
- Department of Gerontology, Shibei Hospital of Jing’an District, Shanghai, 200443, China
| | - Haiya Wang
- Department of Gerontology, Shanghai Ninth People’s Hospital, Shanghai Jiaotong University School of Medicine, Shanghai, 200023, China
| | - Li Jin
- Department of Gerontology, Shibei Hospital of Jing’an District, Shanghai, 200443, China
| |
Collapse
|