Sign in to use this feature.

Years

Between: -

Subjects

remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline

Journals

Article Types

Countries / Regions

Search Results (87)

Search Parameters:
Keywords = matricellular protein

Order results
Result details
Results per page
Select all
Export citation of selected articles as:
16 pages, 1790 KiB  
Review
CYR61 as a Potential Biomarker and Target in Cancer Prognosis and Therapies
by Andrew J. Schenker and Greisha L. Ortiz-Hernández
Cells 2025, 14(11), 761; https://doi.org/10.3390/cells14110761 - 22 May 2025
Viewed by 404
Abstract
Cysteine-rich protein 61 (CYR61) is a matricellular protein in the CCN family that is involved in cellular adhesion, migration, proliferation, and angiogenesis. CYR61 interacts with integrins α6β1, αvβ3, αvβ5, and αIIbβ3 to modulate tumor progression and metastasis while modifying the tumor microenvironment. CYR61 [...] Read more.
Cysteine-rich protein 61 (CYR61) is a matricellular protein in the CCN family that is involved in cellular adhesion, migration, proliferation, and angiogenesis. CYR61 interacts with integrins α6β1, αvβ3, αvβ5, and αIIbβ3 to modulate tumor progression and metastasis while modifying the tumor microenvironment. CYR61 exhibits context-dependent roles in cancer, acting as both a tumor promoter and suppressor. Increased CYR61 expression is linked to extracellular matrix remodeling, immune modulation, and integrin-mediated signaling, making it a potential prognostic biomarker and therapeutic target. Emerging research highlights the utility of CYR61 in liquid biopsies for cancer detection and monitoring. Integrin-targeted therapies, including CYR61-blocking antibodies and CAR-T approaches, offer novel treatment strategies. However, therapy-induced toxicity and resistance remain challenges with these strategies. The further elucidation of the molecular mechanisms of CYR61 may enhance targeted therapeutic interventions and improve patient outcomes. Full article
Show Figures

Graphical abstract

28 pages, 6041 KiB  
Article
CCN2 Activates Cellular Senescence Leading to Kidney Fibrosis in Folic Acid-Induced Experimental Nephropathy
by Lucia Tejedor-Santamaria, Laura Marquez-Exposito, Alicia Villacampa, Vanessa Marchant, Antonio Battaglia-Vieni, Sandra Rayego-Mateos, Raul R. Rodrigues-Diez, Fatima Milhano Santos, Floris A. Valentijn, Sebastian N. Knoppert, Roel Broekhuizen, María Piedad Ruiz-Torres, Roel Goldschmeding, Alberto Ortiz, Concepción Peiró, Tri Q. Nguyen, Adrián M. Ramos and Marta Ruiz-Ortega
Int. J. Mol. Sci. 2025, 26(9), 4401; https://doi.org/10.3390/ijms26094401 - 6 May 2025
Viewed by 395
Abstract
Cellular communication network factor 2 (CCN2, also known as CTGF) is a complex protein that regulates numerous cellular functions. This biomolecule exhibits dual functions, depending on the context, and can act as a matricellular protein or as a growth factor. CCN2 is an [...] Read more.
Cellular communication network factor 2 (CCN2, also known as CTGF) is a complex protein that regulates numerous cellular functions. This biomolecule exhibits dual functions, depending on the context, and can act as a matricellular protein or as a growth factor. CCN2 is an established marker of fibrosis and a well-known mediator of kidney damage, involved in the regulation of inflammation, extracellular matrix remodeling, cell death, and activation of tubular epithelial cell (TECs) senescence. In response to kidney damage, cellular senescence mechanisms are activated, linked to regeneration failure and progression to fibrosis. Our preclinical studies using a total conditional CCN2 knockout mouse demonstrate that CCN2 plays a significant role in the development of a senescence phenotype after exposure to a nephrotoxic agent. CCN2 induces cell growth arrest in TECs, both in the early phase and in the chronic phase of folic acid nephropathy (FAN), associated with cell-death/necroinflammation and fibrosis, respectively. Renal CCN2 overexpression was found to be linked to excessive collagen accumulation in tubulointerstitial areas, microvascular rarefaction, and a decline in renal function, which were observed three weeks following the initial injury. All these findings were markedly diminished in conditional CCN2 knockout mice. In the FAN model, injured senescent TECs are associated with microvascular rarefaction, and both were modulated by CCN2. In primary cultured endothelial cells, as previously described in TECs, CCN2 directly induced senescence. The findings collectively demonstrate the complexity of CCN2, highlight the pivotal role of cellular senescence as an important mechanism in renal injury, and underscore the critical function of this biomolecule in kidney damage progression. Full article
(This article belongs to the Section Molecular Pathology, Diagnostics, and Therapeutics)
Show Figures

Figure 1

21 pages, 9787 KiB  
Article
Pathological Changes in Extracellular Matrix Composition Orchestrate the Fibrotic Feedback Loop Through Macrophage Activation in Dupuytren’s Contracture
by Elizabeth Heinmäe, Kristina Mäemets-Allas, Katre Maasalu, Darja Vastšjonok and Mariliis Klaas
Int. J. Mol. Sci. 2025, 26(7), 3146; https://doi.org/10.3390/ijms26073146 - 28 Mar 2025
Viewed by 1470
Abstract
Dupuytren’s contracture belongs to a group of fibrotic diseases that have similar mechanisms but lack effective treatment and prevention options. The excessive accumulation of connective tissue in Dupuytren’s disease leads to palmar fibrosis that results in contracture deformities. The present study aimed to [...] Read more.
Dupuytren’s contracture belongs to a group of fibrotic diseases that have similar mechanisms but lack effective treatment and prevention options. The excessive accumulation of connective tissue in Dupuytren’s disease leads to palmar fibrosis that results in contracture deformities. The present study aimed to investigate how the tissue microenvironment in Dupuytren’s contracture affects the phenotypic differentiation of macrophages, which leads to an inflammatory response and the development of chronicity in fibrotic disease. We utilized a decellularization-based method combined with proteomic analysis to identify shifts in extracellular matrix composition and the surrounding tissue microenvironment. We found that the expression of several matricellular proteins, such as MFAP4, EFEMP1 (fibulin-3), and ANGPTL2, was elevated in Dupuytren’s tissue. We show that, in response to the changes in the extracellular matrix of Dupuytren’s contracture, macrophages regulate the fibrotic process by cytokine production, promote myofibroblast differentiation, and increase the fibroblast migration rate. Moreover, we found that the extracellular matrix of Dupuytren’s contracture directly supports the macrophage-to-myofibroblast transition, which could be another contributor to Dupuytren’s disease pathogenesis. Our results suggest that interactions between macrophages and the extracellular matrix should be considered as targets for novel fibrotic disease treatment and prevention strategies in the future. Full article
(This article belongs to the Special Issue New Advances in Proteomics in Disease)
Show Figures

Figure 1

17 pages, 5563 KiB  
Article
Increased Plasma Levels of Thrombin-Cleaved Osteopontin in Patients with Delayed Cerebral Infarction After Aneurysmal Subarachnoid Hemorrhage
by Kazuaki Aoki, Fumihiro Kawakita, Koichi Hakozaki, Hideki Kanamaru, Reona Asada, Hidenori Suzuki and pSEED Group
Int. J. Mol. Sci. 2025, 26(6), 2781; https://doi.org/10.3390/ijms26062781 - 19 Mar 2025
Viewed by 349
Abstract
Osteopontin (OPN), a matricellular protein, is produced as a full-length OPN (FL-OPN) and cleaved by thrombin, thus generating the N-terminal half of OPN (OPN N-half) with new functions. Although plasma FL-OPN levels have been associated with neurovascular events after aneurysmal subarachnoid hemorrhage (SAH), [...] Read more.
Osteopontin (OPN), a matricellular protein, is produced as a full-length OPN (FL-OPN) and cleaved by thrombin, thus generating the N-terminal half of OPN (OPN N-half) with new functions. Although plasma FL-OPN levels have been associated with neurovascular events after aneurysmal subarachnoid hemorrhage (SAH), plasma OPN N-half levels have never been investigated. In this study, prospective clinical data and plasma samples were collected from 108 consecutive SAH patients with ruptured aneurysms undergoing acute treatment via surgery, and FL-OPN and OPN N-half levels were measured in plasma with a particular focus on delayed cerebral infarction (DCIn), which has the greatest impact on outcomes. Plasma FL-OPN and OPN N-half levels were intercorrelated and significantly higher in patients with DCIn at days 10–12 post-SAH; a greater area under the receiver-operating characteristic curve was observed for OPN N-half levels, with a cut-off value of 70.42 pmol/L. Multivariate analyses revealed that plasma OPN N-half levels of ≥70.42 pmol/L at days 10–12 were independently associated with DCIn development (adjusted odds ratio, 5.65; 95% confidence interval, 1.68–18.97; p = 0.005). Based on the findings of this study and previous reports, an increase in the OPN N-half level may be indicative of a protective mechanism against DCIn development, and, thus, it holds promise as a new therapeutic target against DCIn after aneurysmal SAH. Full article
Show Figures

Figure 1

12 pages, 1342 KiB  
Article
Periostin from Tumor Stromal Cells Might Be Associated with Malignant Progression of Colorectal Cancer via Smad2/3
by Canfeng Fan, Qiang Wang, Saki Kanei, Kyoka Kawabata, Hinano Nishikubo, Rika Aoyama, Zhonglin Zhu, Daiki Imanishi, Takashi Sakuma, Koji Maruo, Gen Tsujio, Yurie Yamamoto, Tatsunari Fukuoka and Masakazu Yashiro
Cancers 2025, 17(3), 551; https://doi.org/10.3390/cancers17030551 - 6 Feb 2025
Viewed by 827
Abstract
Background/Objectives: Cancer-associated fibroblasts (CAFs) in the tumor microenvironment have been reported to be closely associated with tumor progression in various types of cancer, including colorectal cancer (CRC). Periostin, a matricellular protein, was reported to be expressed on both cancer cells and surrounding tumor [...] Read more.
Background/Objectives: Cancer-associated fibroblasts (CAFs) in the tumor microenvironment have been reported to be closely associated with tumor progression in various types of cancer, including colorectal cancer (CRC). Periostin, a matricellular protein, was reported to be expressed on both cancer cells and surrounding tumor stromal cells, such as CAFs, and is regulated by Smad2/3 signaling. In this study, we aimed to clarify the clinicopathologic significance of periostin and Smad2/3 expression in CRC, with a particular focus on the tumor microenvironment. Methods: A total of 351 CRC patients were enrolled according to the inclusion and exclusion criteria. The expressions of periostin and Smad2/3 in the tumor specimens were examined by immunohistochemistry. Results: Periostin expression of CAFs and cancer cells in the 351 CRC cases was observed at 36.8% and 0.6%, respectively. Smad2/3 expression of CAFs and cancer cells was observed in 41.0% and 90.0%, respectively. In CAFs, high periostin expression was significantly correlated with high Smad2/3 expression, increased invasion depth, lymph node metastasis, venous invasion, advanced disease stage, and a higher rate of relapse. The prognoses of patients with periostin-positive CAFs were significantly poorer than those with periostin-negative CAFs (p < 0.001). The survival outcomes of stage 3 CRC patients with co-expression of periostin and Smad2/3 were significantly worse compared to those with stage 2 CRC. In the stage 3 group, multivariate analysis revealed that periostin was an independent prognostic factor, while univariate analysis showed that both periostin and Smad2/3 were significantly correlated with poor survival. Conclusions: These findings suggest that periostin is expressed mainly in CAFs in CRC and is correlated with Smad2/3 expression in CAFs. Periostin from CAFs might be associated with the malignant progression of CRC via Smad2/3 signaling. Full article
(This article belongs to the Collection New Treatment for Colorectal Cancer)
Show Figures

Figure 1

26 pages, 9308 KiB  
Article
Periostin Splice Variant Expression in Human Osteoblasts from Osteoporotic Patients and Its Effects on Interleukin-6 and Osteoprotegerin
by Till Kuebart, Lisa Oezel, Beyza Gürsoy, Uwe Maus, Joachim Windolf, Bernd Bittersohl and Vera Grotheer
Int. J. Mol. Sci. 2025, 26(3), 932; https://doi.org/10.3390/ijms26030932 - 23 Jan 2025
Viewed by 828
Abstract
Osteoporosis is an inflammatory disease characterised by low bone mass and quality, resulting in weaker bone strength and fragility fractures. Periostin is a matricellular protein expressed in the periosteum of bone by osteoblasts. It regulates cell recruitment and differentiation in response to fracture [...] Read more.
Osteoporosis is an inflammatory disease characterised by low bone mass and quality, resulting in weaker bone strength and fragility fractures. Periostin is a matricellular protein expressed in the periosteum of bone by osteoblasts. It regulates cell recruitment and differentiation in response to fracture and contributes to extracellular matrix (ECM) formation. The aim of the following study was to determine the splice variants of Periostin expressed in human osteoblasts and Periostin’s function in the pathophysiology of osteoporosis. Osteoblasts isolated from femoral heads from 29 patients with or without osteoporosis were utilised. Periostin splice variants were compared by quantitative real-time polymerase chain reaction (qPCR). Furthermore, the effect of Periostin inhibition on osteoblast differentiation was investigated using alizarin red S staining. Lastly, the interaction of IL-6 and Periostin and their effect on osteoprotegerin (OPG) secretion were analysed with the implantation of enzyme-linked immunosorbent assays (ELISAs). It could be demonstrated that human osteoblasts preferentially express Periostin isoform 4, even if splice variant expression was not altered in osteoporosis conditions, indicating that Periostin’s functions in bone are primarily attributable to this isoform. The inhibition of Periostin resulted in significantly reduced osteoblast differentiation. However, Periostin was secreted in significantly higher amounts in osteoblasts from patients with osteoporosis. Additionally, Periostin significantly reduces OPG secretion and, thereby, rather promotes bone resorption. Furthermore, it could be determined that Periostin and IL-6 induce each other, and both significantly decrease OPG secretion. A positive feedback loop exacerbates the dysregulation found in human osteoblasts from patients with osteoporosis, thereby contributing to bone loss. Full article
(This article belongs to the Special Issue Advances in Osteogenesis)
Show Figures

Figure 1

21 pages, 3287 KiB  
Article
Stage-Dependent Fibrotic Gene Profiling of WISP1-Mediated Fibrogenesis in Human Fibroblasts
by Kirti Singh, Marta Witek, Jaladhi Brahmbhatt, Jacquelyn McEntire, Kannan Thirunavukkarasu and Sunday S. Oladipupo
Cells 2024, 13(23), 2005; https://doi.org/10.3390/cells13232005 - 5 Dec 2024
Cited by 1 | Viewed by 1436
Abstract
Idiopathic pulmonary fibrosis (IPF) is the most common interstitial lung disease with unknown etiology, characterized by chronic inflammation and tissue scarring. Although, Pirfenidone and Nintedanib slow the disease progression, no currently available drugs or therapeutic interventions address the underlying cause, highlighting the unmet [...] Read more.
Idiopathic pulmonary fibrosis (IPF) is the most common interstitial lung disease with unknown etiology, characterized by chronic inflammation and tissue scarring. Although, Pirfenidone and Nintedanib slow the disease progression, no currently available drugs or therapeutic interventions address the underlying cause, highlighting the unmet medical need. A matricellular protein, Wnt-1-induced secreted protein 1 (WISP1), also referred to as CCN4 (cellular communication network factor 4), is a secreted multi-modular protein implicated in multi-organ fibrosis. Although the precise mechanism of WISP1-mediated fibrosis remains unclear, emerging evidence indicates that WISP1 is profibrotic in nature. While WISP1-targeting therapy is applied in the clinic for fibrosis, detailed interrogation of WISP1-mediated fibrogenic molecular and biological pathways is lacking. Here, for the first time, using NanoString® technology, we identified a novel WISP1-associated profibrotic gene signature and molecular pathways potentially involved in the initiation and progression of fibrosis in primary human dermal and lung fibroblasts from both healthy individuals and IPF patients. Our data demonstrate that WISP1 is upregulated in IPF-lung fibroblasts as compared to healthy control. Furthermore, our results confirm that WISP1 is downstream of the transforming growth factor-β (TGFβ), and it induces fibroblast cell proliferation. Additionally, WISP1 induced IL6 and CCL2 in fibroblasts. We also developed a novel, combined TGFβ and WISP1 in vitro system to demonstrate a role for WISP1 in the progression of fibrosis. Overall, our findings uncover not only similarities but also striking differences in the molecular profile of WISP1 in human fibroblasts, both during the initiation and progression phases, as well as in disease-specific context. Full article
Show Figures

Graphical abstract

14 pages, 4068 KiB  
Perspective
The Origins of Engineered Biomaterials: NSF-Funded, University of Washington Engineered Biomaterials (UWEB)
by Buddy D. Ratner
Bioengineering 2024, 11(11), 1117; https://doi.org/10.3390/bioengineering11111117 - 6 Nov 2024
Cited by 1 | Viewed by 1035
Abstract
The University of Washington Engineered Biomaterials (UWEB) Engineering Research Center (ERC) was funded from 1996 to 2007 by the U.S. National Science Foundation. The mission of UWEB was to advance biomaterials by integrating modern biology with materials science. UWEB specifically focused on the [...] Read more.
The University of Washington Engineered Biomaterials (UWEB) Engineering Research Center (ERC) was funded from 1996 to 2007 by the U.S. National Science Foundation. The mission of UWEB was to advance biomaterials by integrating modern biology with materials science. UWEB specifically focused on the healing and integration of medical implants. UWEB teamed biologists, physicians, engineers, and industry and demonstrated three paths that might advance biomaterials so they could seamlessly integrate and heal in the body. The three primary lines of investigation were precision porous scaffolds, super-non-fouling surfaces, and the control of matricellular proteins. The UWEB program set the groundwork for the modern field of immunoengineering. Also, UWEB invested significantly in training scientists/engineers who could freely integrate advances in biological sciences, state-of-the-art materials science, and medical technology. This historical summary of the UWEB program demonstrates that federal investment in interfacing forefront fields can yield dividends with benefits for society and the economy. Full article
(This article belongs to the Special Issue 10th Anniversary of Bioengineering: Perspectives in Bioengineering)
Show Figures

Figure 1

28 pages, 21118 KiB  
Article
Galectin-3/Gelatin Electrospun Scaffolds Modulate Collagen Synthesis in Skin Healing but Do Not Improve Wound Closure Kinetics
by Karrington A. McLeod, Madeleine Di Gregorio, Dylan Tinney, Justin Carmichael, David Zuanazzi, Walter L. Siqueira, Amin Rizkalla and Douglas W. Hamilton
Bioengineering 2024, 11(10), 960; https://doi.org/10.3390/bioengineering11100960 - 25 Sep 2024
Cited by 1 | Viewed by 1430
Abstract
Chronic wounds remain trapped in a pro-inflammatory state, with strategies targeted at inducing re-epithelialization and the proliferative phase of healing desirable. As a member of the lectin family, galectin-3 is implicated in the regulation of macrophage phenotype and epithelial migration. We investigated if [...] Read more.
Chronic wounds remain trapped in a pro-inflammatory state, with strategies targeted at inducing re-epithelialization and the proliferative phase of healing desirable. As a member of the lectin family, galectin-3 is implicated in the regulation of macrophage phenotype and epithelial migration. We investigated if local delivery of galectin-3 enhanced skin healing in a full-thickness excisional C57BL/6 mouse model. An electrospun gelatin scaffold loaded with galectin-3 was developed and compared to topical delivery of galectin-3. Electrospun gelatin/galectin-3 scaffolds had an average fiber diameter of 200 nm, with 83% scaffold porosity approximately and an average pore diameter of 1.15 μm. The developed scaffolds supported dermal fibroblast adhesion, matrix deposition, and proliferation in vitro. In vivo treatment of 6 mm full-thickness excisional wounds with gelatin/galectin-3 scaffolds did not influence wound closure, re-epithelialization, or macrophage phenotypes, but increased collagen synthesis. In comparison, topical delivery of galectin-3 [6.7 µg/mL] significantly increased arginase-I cell density at day 7 versus untreated and gelatin/galectin-3 scaffolds (p < 0.05). A preliminary assessment of increasing the concentration of topical galectin-3 demonstrated that at day 7, galectin-3 [12.5 µg/mL] significantly increased both epithelial migration and collagen content in a concentration-dependent manner. In conclusion, local delivery of galectin 3 shows potential efficacy in modulating skin healing in a concentration-dependent manner. Full article
(This article belongs to the Special Issue Biomaterials and Technology for Skin Wound Healing)
Show Figures

Graphical abstract

14 pages, 3263 KiB  
Article
Ccn2 Deletion Reduces Cardiac Dysfunction, Oxidative Markers, and Fibrosis Induced by Doxorubicin Administration in Mice
by Antonio Tejera-Muñoz, Marcelino Cortés, Alianet Rodriguez-Rodriguez, Lucia Tejedor-Santamaria, Vanessa Marchant, Sandra Rayego-Mateos, Maria José Gimeno-Longas, Andrew Leask, Tri Q. Nguyen, María Martín, Jose Tuñón, Isabel Rodríguez, Marta Ruiz-Ortega and Raul R. Rodrigues-Díez
Int. J. Mol. Sci. 2024, 25(17), 9617; https://doi.org/10.3390/ijms25179617 - 5 Sep 2024
Viewed by 4061
Abstract
Cellular Communication Network Factor 2 (CCN2) is a matricellular protein implicated in cell communication and microenvironmental signaling. Overexpression of CCN2 has been documented in various cardiovascular pathologies, wherein it may exert either deleterious or protective effects depending on the pathological context, thereby suggesting [...] Read more.
Cellular Communication Network Factor 2 (CCN2) is a matricellular protein implicated in cell communication and microenvironmental signaling. Overexpression of CCN2 has been documented in various cardiovascular pathologies, wherein it may exert either deleterious or protective effects depending on the pathological context, thereby suggesting that its role in the cardiovascular system is not yet fully elucidated. In this study, we aimed to investigate the effects of Ccn2 gene deletion on the progression of acute cardiac injury induced by doxorubicin (DOX), a widely utilized chemotherapeutic agent. To this end, we employed conditional knockout (KO) mice for the Ccn2 gene (CCN2-KO), which were administered DOX and compared to DOX-treated wild-type (WT) control mice. Our findings demonstrated that the ablation of CCN2 ameliorated DOX-induced cardiac dysfunction, as evidenced by improvements in ejection fraction (EF) and fractional shortening (FS) of the left ventricle. Furthermore, DOX-treated CCN2-KO mice exhibited a significant reduction in the gene expression and activation of oxidative stress markers (Hmox1 and Nfe2l2/NRF2) relative to DOX-treated WT controls. Additionally, the deletion of Ccn2 markedly attenuated DOX-induced cardiac fibrosis. Collectively, these results suggest that CCN2 plays a pivotal role in the pathogenesis of DOX-mediated cardiotoxicity by modulating oxidative stress and fibrotic pathways. These findings provide a novel avenue for future investigations to explore the therapeutic potential of targeting CCN2 in the prevention of DOX-induced cardiac dysfunction. Full article
(This article belongs to the Section Molecular Biology)
Show Figures

Figure 1

23 pages, 412 KiB  
Review
Involvement of Matricellular Proteins in Cellular Senescence: Potential Therapeutic Targets for Age-Related Diseases
by Motomichi Fujita, Manabu Sasada, Takuya Iyoda and Fumio Fukai
Int. J. Mol. Sci. 2024, 25(12), 6591; https://doi.org/10.3390/ijms25126591 - 15 Jun 2024
Cited by 6 | Viewed by 2078
Abstract
Senescence is a physiological and pathological cellular program triggered by various types of cellular stress. Senescent cells exhibit multiple characteristic changes. Among them, the characteristic flattened and enlarged morphology exhibited in senescent cells is observed regardless of the stimuli causing the senescence. Several [...] Read more.
Senescence is a physiological and pathological cellular program triggered by various types of cellular stress. Senescent cells exhibit multiple characteristic changes. Among them, the characteristic flattened and enlarged morphology exhibited in senescent cells is observed regardless of the stimuli causing the senescence. Several studies have provided important insights into pro-adhesive properties of cellular senescence, suggesting that cell adhesion to the extracellular matrix (ECM), which is involved in characteristic morphological changes, may play pivotal roles in cellular senescence. Matricellular proteins, a group of structurally unrelated ECM molecules that are secreted into the extracellular environment, have the unique ability to control cell adhesion to the ECM by binding to cell adhesion receptors, including integrins. Recent reports have certified that matricellular proteins are closely involved in cellular senescence. Through this biological function, matricellular proteins are thought to play important roles in the pathogenesis of age-related diseases, including fibrosis, osteoarthritis, intervertebral disc degeneration, atherosclerosis, and cancer. This review outlines recent studies on the role of matricellular proteins in inducing cellular senescence. We highlight the role of integrin-mediated signaling in inducing cellular senescence and provide new therapeutic options for age-related diseases targeting matricellular proteins and integrins. Full article
(This article belongs to the Section Bioactives and Nutraceuticals)
14 pages, 8408 KiB  
Article
Modified mRNA-Mediated CCN5 Gene Transfer Ameliorates Cardiac Dysfunction and Fibrosis without Adverse Structural Remodeling
by Min Ho Song, Jimeen Yoo, Do-A Kwon, Elena Chepurko, Sunghye Cho, Anthony Fargnoli, Roger J. Hajjar, Woo Jin Park, Lior Zangi and Dongtak Jeong
Int. J. Mol. Sci. 2024, 25(11), 6262; https://doi.org/10.3390/ijms25116262 - 6 Jun 2024
Cited by 3 | Viewed by 1847
Abstract
Modified mRNAs (modRNAs) are an emerging delivery method for gene therapy. The success of modRNA-based COVID-19 vaccines has demonstrated that modRNA is a safe and effective therapeutic tool. Moreover, modRNA has the potential to treat various human diseases, including cardiac dysfunction. Acute myocardial [...] Read more.
Modified mRNAs (modRNAs) are an emerging delivery method for gene therapy. The success of modRNA-based COVID-19 vaccines has demonstrated that modRNA is a safe and effective therapeutic tool. Moreover, modRNA has the potential to treat various human diseases, including cardiac dysfunction. Acute myocardial infarction (MI) is a major cardiac disorder that currently lacks curative treatment options, and MI is commonly accompanied by fibrosis and impaired cardiac function. Our group previously demonstrated that the matricellular protein CCN5 inhibits cardiac fibrosis (CF) and mitigates cardiac dysfunction. However, it remains unclear whether early intervention of CF under stress conditions is beneficial or more detrimental due to potential adverse effects such as left ventricular (LV) rupture. We hypothesized that CCN5 would alleviate the adverse effects of myocardial infarction (MI) through its anti-fibrotic properties under stress conditions. To induce the rapid expression of CCN5, ModRNA-CCN5 was synthesized and administrated directly into the myocardium in a mouse MI model. To evaluate CCN5 activity, we established two independent experimental schemes: (1) preventive intervention and (2) therapeutic intervention. Functional analyses, including echocardiography and magnetic resonance imaging (MRI), along with molecular assays, demonstrated that modRNA-mediated CCN5 gene transfer significantly attenuated cardiac fibrosis and improved cardiac function in both preventive and therapeutic models, without causing left ventricular rupture or any adverse cardiac remodeling. In conclusion, early intervention in CF by ModRNA-CCN5 gene transfer is an efficient and safe therapeutic modality for treating MI-induced heart failure. Full article
(This article belongs to the Special Issue Calcium Homeostasis of Cells in Health and Disease)
Show Figures

Figure 1

26 pages, 5445 KiB  
Article
Myocardin-Related Transcription Factor Mediates Epithelial Fibrogenesis in Polycystic Kidney Disease
by Zsuzsanna Lichner, Mei Ding, Tarang Khare, Qinghong Dan, Raquel Benitez, Mercédesz Praszner, Xuewen Song, Rola Saleeb, Boris Hinz, York Pei, Katalin Szászi and András Kapus
Cells 2024, 13(11), 984; https://doi.org/10.3390/cells13110984 - 5 Jun 2024
Cited by 1 | Viewed by 1663
Abstract
Polycystic kidney disease (PKD) is characterized by extensive cyst formation and progressive fibrosis. However, the molecular mechanisms whereby the loss/loss-of-function of Polycystin 1 or 2 (PC1/2) provokes fibrosis are largely unknown. The small GTPase RhoA has been recently implicated in cystogenesis, and [...] Read more.
Polycystic kidney disease (PKD) is characterized by extensive cyst formation and progressive fibrosis. However, the molecular mechanisms whereby the loss/loss-of-function of Polycystin 1 or 2 (PC1/2) provokes fibrosis are largely unknown. The small GTPase RhoA has been recently implicated in cystogenesis, and we identified the RhoA/cytoskeleton/myocardin-related transcription factor (MRTF) pathway as an emerging mediator of epithelium-induced fibrogenesis. Therefore, we hypothesized that MRTF is activated by PC1/2 loss and plays a critical role in the fibrogenic reprogramming of the epithelium. The loss of PC1 or PC2, induced by siRNA in vitro, activated RhoA and caused cytoskeletal remodeling and robust nuclear MRTF translocation and overexpression. These phenomena were also manifested in PKD1 (RC/RC) and PKD2 (WS25/−) mice, with MRTF translocation and overexpression occurring predominantly in dilated tubules and the cyst-lining epithelium, respectively. In epithelial cells, a large cohort of PC1/PC2 downregulation-induced genes was MRTF-dependent, including cytoskeletal, integrin-related, and matricellular/fibrogenic proteins. Epithelial MRTF was necessary for the paracrine priming of the fibroblast–myofibroblast transition. Thus, MRTF acts as a prime inducer of epithelial fibrogenesis in PKD. We propose that RhoA is a common upstream inducer of both histological hallmarks of PKD: cystogenesis and fibrosis. Full article
Show Figures

Graphical abstract

16 pages, 2851 KiB  
Article
Dynamics of Matricellular Protein Levels in Blood Predict Recovery in Patients with Human Immunodeficiency Virus-Tuberculosis Coinfection
by Ashwini Shete, Manisha Ghate, Hiroko Iwasaki-Hozumi, Sandip Patil, Pallavi Shidhaye, Gaowa Bai, Takashi Matsuba, Pratiksha Pharande, Bharati Mahajan, Aarti Randive, Anupam Mukherjee and Toshio Hattori
Viruses 2024, 16(5), 664; https://doi.org/10.3390/v16050664 - 24 Apr 2024
Cited by 2 | Viewed by 1681
Abstract
Chronic immune activation in tuberculosis (TB) associated with human immunodeficiency virus (HIV) infection (HIV/TB) modifies their clinical course. We prospectively measured osteopontin (OPN), full-length galectin-9 (FL-Gal9), and total-Gal9 (T-Gal9) levels in 32 patients with HIV/TB coinfection treated with anti-tuberculosis and antiretroviral therapies over [...] Read more.
Chronic immune activation in tuberculosis (TB) associated with human immunodeficiency virus (HIV) infection (HIV/TB) modifies their clinical course. We prospectively measured osteopontin (OPN), full-length galectin-9 (FL-Gal9), and total-Gal9 (T-Gal9) levels in 32 patients with HIV/TB coinfection treated with anti-tuberculosis and antiretroviral therapies over 6–18 months to determine the amelioration of inflammatory conditions in response to the therapies. We observed a significant time-dependent decrease in FL-Gal9 in both pulmonary TB (PTB, n = 20) and extrapulmonary TB (EPTB, n = 12) patients. The levels of T-Gal9, OPN, and CRP decreased significantly after treatment in only PTB patients. We calculated the inflammatory score (INS) indicating immunologic recovery based on the decline in OPN, FL-Gal9, T-Gal9, and CRP levels. Baseline levels of T-Gal9 and OPN positively correlated with INS in all TB and only PTB patients, respectively, indicating that their levels predict better recovery. In contrast, FL-Gal9 levels at the second visit negatively correlated with INS in EPTB patients. The decrease rate in OPN levels at the second visit also correlated positively with INS in PTB patients. Women showed a higher INS and lower levels of FL-Gal9 than men. The patients with moderate grade severity on chest X-ray had higher CD4 cell numbers than those with limited grade severity. Monitoring these markers will help to predict and assess the response to therapy as well as to devise strategies to reduce the complications caused by chronic immune activation in patients with HIV/TB coinfection. Full article
(This article belongs to the Special Issue Tuberculosis (TB) and HIV Coinfection)
Show Figures

Figure 1

17 pages, 3694 KiB  
Article
Global Proteomics Analysis of Lysophosphatidic Acid Signaling in PC-3 Human Prostate Cancer Cells: Role of CCN1
by Pravita Balijepalli, Guihua Yue, Bhagwat Prasad and Kathryn E. Meier
Int. J. Mol. Sci. 2024, 25(4), 2067; https://doi.org/10.3390/ijms25042067 - 8 Feb 2024
Cited by 1 | Viewed by 1934
Abstract
Cysteine-rich angiogenic factor 61 (CCN1/Cyr61) is a matricellular protein that is induced and secreted in response to growth factors. Our previous work showed that 18:1-lysophosphatidic acid (LPA), which activates the G protein-coupled receptor LPAR1, induces CCN1 between 2–4 h in PC-3 human prostate [...] Read more.
Cysteine-rich angiogenic factor 61 (CCN1/Cyr61) is a matricellular protein that is induced and secreted in response to growth factors. Our previous work showed that 18:1-lysophosphatidic acid (LPA), which activates the G protein-coupled receptor LPAR1, induces CCN1 between 2–4 h in PC-3 human prostate cancer cells in a manner than enhances cell-substrate adhesion. While the time course of induction suggests that CCN1 contributes to intermediate events in LPA action, the roles of CCN1 in LPA-mediated signal transduction have not been fully elucidated. This study utilized a comprehensive global proteomics approach to identify proteins up- or down-regulated in response to treatment of PC-3 cells with LPA for three hours, during the time of peak CCN1 levels. In addition, the effects of siRNA-mediated CCN1 knockdown on LPA responses were analyzed. The results show that, in addition to CCN1, LPA increased the levels of multiple proteins. Proteins up-regulated by LPA included metastasis-associated in colon cancer protein 1 (MACC1) and thrombospondin-1 (TSP1/THBS1); both MACC1 and TSP1 regulated cancer cell adhesion and motility. LPA down-regulated thioredoxin interacting protein (TXNIP). CCN1 knockdown suppressed the LPA-induced up-regulation of 30 proteins; these included MACC1 and TSP1, as confirmed by immunoblotting. Gene ontology and STRING analyses revealed multiple pathways impacted by LPA and CCN1. These results indicate that CCN1 contributes to LPA signaling cascades that occur during the intermediate phase after the initial stimulus. The study provides a rationale for the development of interventions to disrupt the LPA-CCN1 axis. Full article
(This article belongs to the Special Issue Lysophosphatidic Acid Signaling in Health and Disease)
Show Figures

Figure 1

Back to TopTop