Sign in to use this feature.

Years

Between: -

Subjects

remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline

Journals

Article Types

Countries / Regions

Search Results (112)

Search Parameters:
Keywords = recombinant human collagen

Order results
Result details
Results per page
Select all
Export citation of selected articles as:
19 pages, 6032 KiB  
Article
Recombinant Human Annexin A5 Ameliorates Localized Scleroderma by Inhibiting the Activation of Fibroblasts and Macrophages
by Bijun Kang, Zhuoxuan Jia, Wei Li and Wenjie Zhang
Pharmaceutics 2025, 17(8), 986; https://doi.org/10.3390/pharmaceutics17080986 (registering DOI) - 30 Jul 2025
Viewed by 116
Abstract
Background: Localized scleroderma (LoS) is a chronic autoimmune condition marked by cutaneous fibrosis and persistent inflammation. Modulating the activation of inflammatory cells and fibroblasts remains a central strategy in LoS treatment. We investigate the anti-fibrotic effects of Annexin A5 (AnxA5), identified as [...] Read more.
Background: Localized scleroderma (LoS) is a chronic autoimmune condition marked by cutaneous fibrosis and persistent inflammation. Modulating the activation of inflammatory cells and fibroblasts remains a central strategy in LoS treatment. We investigate the anti-fibrotic effects of Annexin A5 (AnxA5), identified as a key inflammatory component in fat extract, and assess its therapeutic efficacy. Methods: In vitro experiments were performed using TGF-β-stimulated primary human dermal fibroblasts treated with recombinant AnxA5. The anti-fibrotic effects and underlying mechanisms were assessed using CCK-8 assays, quantitative real-time PCR, Western blotting, and immunocytochemistry. In vivo, AnxA5 was administered via both preventative and therapeutic protocols in bleomycin-induced LoS mouse models. Treatment outcomes were evaluated by histological staining, collagen quantification, immunostaining, and measurement of pro-inflammatory cytokines. Results: TGF-β stimulation induced myofibroblast differentiation and extracellular matrix (ECM) production in dermal fibroblasts, both of which were significantly attenuated by AnxA5 treatment through the inhibition of phosphorylation of Smad2. In vivo, both preventative and therapeutic administration of AnxA5 effectively reduced dermal thickness, collagen deposition, ECM accumulation, M1 macrophage infiltration, and levels of pro-inflammatory cytokines. Conclusions: Through both preventative and therapeutic administration, AnxA5 ameliorates LoS by exerting dual anti-fibrotic and anti-inflammatory effects, underscoring its potential for treating fibrotic diseases. Full article
(This article belongs to the Section Biopharmaceutics)
Show Figures

Figure 1

18 pages, 8559 KiB  
Article
Recombinant Type XVII Collagen Promotes Hair Growth by Activating the Wnt/β-Catenin and SHH/GLI Signaling Pathways
by Yuyao Zhang, Shiyu Yin, Ru Xu, Jiayu Xiao, Rui Yi, Jiahui Mao, Zhiguang Duan and Daidi Fan
Cosmetics 2025, 12(4), 156; https://doi.org/10.3390/cosmetics12040156 - 23 Jul 2025
Viewed by 524
Abstract
(1) Background: As society progresses, increasing numbers of individuals are experiencing hair loss, which can be attributed to factors such as unhealthy diets, insufficient sleep, stress, and hormonal imbalances. Currently available pharmacological treatments for hair loss often cause undesirable side effects, highlighting the [...] Read more.
(1) Background: As society progresses, increasing numbers of individuals are experiencing hair loss, which can be attributed to factors such as unhealthy diets, insufficient sleep, stress, and hormonal imbalances. Currently available pharmacological treatments for hair loss often cause undesirable side effects, highlighting the urgent need to explore safer and more effective agents to promote hair restoration. This study investigated the role of recombinant human type XVII collagen derived from the α1 chain (rhCOL17A1) in facilitating hair growth and restoration. (2) Methods: We analyzed the impact of rhCOL17A1 on the mRNA expression of several growth factors, as well as Bcl-2 and Bax, at the cellular level. Moreover, the effects of rhCOL17A1 on the expression of key proteins in the Wnt/β-catenin and Sonic Hedgehog (SHH)/GLI signaling pathways were examined by Western blotting (WB). At the organismal level, we established a model in C57BL/6 mice through chronic subcutaneous administration of 5% testosterone propionate. We subsequently assessed the effect of rhCOL17A1 on hair regrowth via histological analysis using hematoxylin and eosin (H&E) staining and immunofluorescence staining. (3) Results: rhCOL17A1 contributes to the resistance of hair follicle dermal papilla cells (HFDPCs) to apoptosis. rhCOL17A1 activates the Wnt/β-catenin and SHH/GLI signaling pathways, and increases the expression of type XVII collagen (COLXVII), thereby creating a favorable environment for hair growth. Furthermore, rhCOL17A1 exerts a significant growth-promoting effect at the animal level. (4) Conclusions: rhCOL17 promotes hair growth by activating the Wnt/β-catenin and SHH/GLI signaling pathways and upregulating COLXVII expression. Full article
Show Figures

Figure 1

31 pages, 865 KiB  
Review
Sustainable Hydrogels for Medical Applications: Biotechnological Innovations Supporting One Health
by Silvia Romano, Sorur Yazdanpanah, Orsolina Petillo, Raffaele Conte, Fabrizia Sepe, Gianfranco Peluso and Anna Calarco
Gels 2025, 11(7), 559; https://doi.org/10.3390/gels11070559 - 21 Jul 2025
Viewed by 454
Abstract
The One Health paradigm—recognizing the interconnected health of humans, animals, and the environment—promotes the development of sustainable technologies that enhance human health while minimizing ecological impact. In this context, bio-based hydrogels have emerged as a promising class of biomaterials for advanced medical applications. [...] Read more.
The One Health paradigm—recognizing the interconnected health of humans, animals, and the environment—promotes the development of sustainable technologies that enhance human health while minimizing ecological impact. In this context, bio-based hydrogels have emerged as a promising class of biomaterials for advanced medical applications. Produced through biotechnological methods such as genetic engineering and microbial fermentation, these hydrogels are composed of renewable and biocompatible materials, including recombinant collagen, elastin, silk fibroin, bacterial cellulose, xanthan gum, and hyaluronic acid. Their high water content, structural tunability, and biodegradability make them ideal candidates for various biomedical applications such as wound healing, tissue regeneration, and the design of extracellular matrix (ECM)-mimicking scaffolds. By offering controlled mechanical properties, biocompatibility, and the potential for minimally invasive administration, sustainable hydrogels represent a strategic innovation for regenerative medicine and therapeutic interventions. This review discusses the characteristics and medical applications of these hydrogels, highlighting their role in advancing sustainable healthcare solutions within the One Health framework. Full article
(This article belongs to the Special Issue Application of Hydrogels in Medicine)
Show Figures

Figure 1

15 pages, 526 KiB  
Review
Advancements in Clinical Utilization of Recombinant Human Collagen: An Extensive Review
by Isaac Wong Kai Jie, Kar Wai Alvin Lee, Song Eun Yoon, Jong Keun Song, Lisa Kwin Wah Chan, Cheuk Hung Lee, Eunji Jeong, Jin-Hyun Kim and Kyu-Ho Yi
Life 2025, 15(4), 582; https://doi.org/10.3390/life15040582 - 1 Apr 2025
Cited by 3 | Viewed by 2809
Abstract
Introduction: Recombinant human collagen, developed through advanced recombinant DNA technology, has emerged as a cutting-edge biomaterial with diverse applications in medicine. It addresses significant limitations of animal-derived collagens, such as immunogenicity and the risk of zoonotic diseases. Objective: This review evaluates the clinical [...] Read more.
Introduction: Recombinant human collagen, developed through advanced recombinant DNA technology, has emerged as a cutting-edge biomaterial with diverse applications in medicine. It addresses significant limitations of animal-derived collagens, such as immunogenicity and the risk of zoonotic diseases. Objective: This review evaluates the clinical applications, benefits, and challenges associated with recombinant human collagen, focusing on its potential to transform medical and surgical practices. Methods: A comprehensive search was conducted in MEDLINE, PubMed, and Ovid databases using keywords such as “Recombinant Human Collagen”, “Collagen-Based Biomaterials”, “Clinical Applications”, “Tissue Repair”, and “Wound Healing”. Relevant studies, including clinical trials and diagnostic applications, were analyzed and classified according to the Oxford Centre for Evidence-Based Medicine evidence hierarchy. Findings: Recombinant human collagen demonstrates superior mechanical properties and controlled degradation rates compared to traditional collagen sources. Clinical studies highlight its effectiveness in accelerating wound closure, promoting dermal regeneration, and minimizing scarring, making it particularly valuable in chronic wound management and surgical interventions. In tissue engineering, recombinant human collagen scaffolds have shown potential for regenerating cartilage, bone, and cardiovascular tissues by supporting cell proliferation, differentiation, and matrix deposition. Additionally, its adaptability for forming hydrogels and matrices enhances its suitability for drug delivery systems, enabling controlled and sustained release of therapeutic agents. Conclusion: Recombinant human collagen represents a transformative advancement in clinical practice, providing a safer and more effective alternative to traditional collagen sources. Its demonstrated success in wound healing, tissue engineering, and drug delivery highlights its potential to significantly improve patient outcomes. However, challenges such as high production costs, regulatory complexities, and long-term biocompatibility remain barriers to widespread clinical adoption. Further research and collaboration between biotechnology developers and regulatory authorities are essential to fully realize its clinical potential. Full article
Show Figures

Figure 1

38 pages, 1377 KiB  
Review
Advances in Molecular Function and Recombinant Expression of Human Collagen
by Wenli Sun, Mohamad Hesam Shahrajabian, Kun Ma and Shubin Wang
Pharmaceuticals 2025, 18(3), 430; https://doi.org/10.3390/ph18030430 - 18 Mar 2025
Cited by 3 | Viewed by 2406
Abstract
Collagen is the main protein found in skin, bone, cartilage, ligaments, tendons and connective tissue, and it can exhibit properties ranging from compliant to rigid or form gradients between these states. The collagen family comprises 28 members, each containing at least one triple-helical [...] Read more.
Collagen is the main protein found in skin, bone, cartilage, ligaments, tendons and connective tissue, and it can exhibit properties ranging from compliant to rigid or form gradients between these states. The collagen family comprises 28 members, each containing at least one triple-helical domain. These proteins play critical roles in maintaining mechanical characteristics, tissue organization, and structural integrity. Collagens regulate cellular processes such as proliferation, migration, and differentiation through interactions with cell surface receptors. Fibrillar collagens, the most abundant extracellular matrix (ECM) proteins, provide organs and tissues with structural stability and connectivity. In the mammalian myocardial interstitium, types I and III collagens are predominant: collagen I is found in organs, tendons, and bones; collagen II is found in cartilage; collagen III is found in reticular fibers; collagen IV is found in basement membranes; and collagen V is found in nails and hair. Recombinant human collagens, particularly in sponge-like porous formats combined with bone morphogenetic proteins, serve as effective scaffolds for bone repair. Due to their biocompatibility and low immunogenicity, collagens are pivotal in tissue engineering applications for skin, bone, and wound regeneration. Recombinant technology enables the production of triple-helical collagens with amino acid sequences identical to human tissue-derived collagens. This review summarizes recent advances in the molecular functions and recombinant expression of human collagens, with a focus on their biomedical applications. Full article
Show Figures

Figure 1

20 pages, 13597 KiB  
Article
Suture-Mediated Delivery System Reduces the Incidence of Uterine Scarring Through the TGF-β Pathway
by He Bai, Wei Zhang, Xuanxuan Yan, Lin Qiu, Pengfei Cui and Weiyang Chen
J. Funct. Biomater. 2025, 16(2), 52; https://doi.org/10.3390/jfb16020052 - 7 Feb 2025
Viewed by 1258
Abstract
In recent years, factors such as the postponement of childbearing and the relaxation of the childbearing policy have led to an increase in the proportion of cesarean sections and other intrauterine surgeries among pregnant women, further increasing the incidence of uterine scars. Currently, [...] Read more.
In recent years, factors such as the postponement of childbearing and the relaxation of the childbearing policy have led to an increase in the proportion of cesarean sections and other intrauterine surgeries among pregnant women, further increasing the incidence of uterine scars. Currently, there is a lack of effective clinical treatment methods for uterine scars. In this study, a suture loaded with gene medicine was designed for the repair of uterine scars. Specifically, the non-viral vector Lipo8000 was first used to form a complex solution with the plasmid TGF-β3. Then, it was mixed and adsorbed with the surgical sutures pretreated with recombinant human type III collagen (RhCol III). In vitro experiments confirmed that RhCol III and the plasmid were successfully loaded onto the sutures and could be released and expressed. In vivo experiments were carried out using a rat model simulating uterine scars. The section results showed that compared with the scar model group, the expression level of TGF-β3 in the RhCol III+TGF-β3 group increased by 39%, the expression level of TGF-β1 decreased by 62.8%, and the fibrosis rate decreased by 16.8%, which has a positive effect on the prevention of uterine scars. This study integrates the therapeutic medicine into the sutures, ensuring that the medicine can come into contact with the wound site after suturing. Moreover, RhCol III and the gene medicine work synergistically to promote the repair of uterine wounds. Full article
Show Figures

Figure 1

18 pages, 27979 KiB  
Article
Recombinant Humanized Collagen Enhances Secreted Protein Levels of Fibroblasts and Facilitates Rats’ Skin Basement Membrane Reinforcement
by Shijia Ye, Boyu Chen, Lakshmi Jeevithan, Haoze Yang, Yaqi Kong, Xiaozhen Diao and Wenhui Wu
J. Funct. Biomater. 2025, 16(2), 47; https://doi.org/10.3390/jfb16020047 - 1 Feb 2025
Viewed by 1443
Abstract
Collagen and its peptides exhibit remarkable antioxidant activity, superior biocompatibility, and water solubility, making them a significant research focus in skin care. Hence, the recombinant humanized collagen types I, III, and XVII complexed with niacinamide were developed to address damage in human foreskin [...] Read more.
Collagen and its peptides exhibit remarkable antioxidant activity, superior biocompatibility, and water solubility, making them a significant research focus in skin care. Hence, the recombinant humanized collagen types I, III, and XVII complexed with niacinamide were developed to address damage in human foreskin fibroblasts (HFF-1) caused by ultraviolet radiation and to evaluate basement membrane proteins in a rat skin model. The Cell Counting Kit-8 (CCK-8) assay showed that higher concentrations of the complex increased the survival of damaged cells by approximately 10% and 22%, respectively, compared to the normal group after 16 and 48 h of treatment. Further biochemical analyses using ELISA and immunofluorescence (IF) confirmed that the complex enhanced the expression of collagen type IV, laminin, P63, and transforming growth factor-β (TGF-β) in the damaged cells. Additionally, the complex boosted the activity of the basement membrane in rat skin and stimulated the secretion of integrin, laminin, and perlecan. Overall, the recombinant humanized collagen complex effectively reinforced the skin’s basement membrane. Full article
Show Figures

Figure 1

14 pages, 7247 KiB  
Article
Development of Recombinant Human Collagen-Based Porous Scaffolds for Skin Tissue Engineering: Enhanced Mechanical Strength and Biocompatibility
by Yang Yang, Ting Yu, Mengdan Tao, Yong Wang, Xinying Yao, Chenkai Zhu, Fengxue Xin and Min Jiang
Polymers 2025, 17(3), 303; https://doi.org/10.3390/polym17030303 - 23 Jan 2025
Cited by 1 | Viewed by 1592
Abstract
Skin tissue engineering scaffolds should possess key properties such as porosity, degradability, durability, and biocompatibility to effectively facilitate skin cell adhesion and growth. In this study, recombinant human collagen (RHC) was used to fabricate porous scaffolds via freeze-drying, offering an alternative to animal-derived [...] Read more.
Skin tissue engineering scaffolds should possess key properties such as porosity, degradability, durability, and biocompatibility to effectively facilitate skin cell adhesion and growth. In this study, recombinant human collagen (RHC) was used to fabricate porous scaffolds via freeze-drying, offering an alternative to animal-derived collagen where bovine collagen (BC)-based scaffolds were also prepared for comparison. The internal morphology of the RHC scaffolds were characterized by scanning electron microscopy (SEM) and the pore size ranged from 68.39 to 117.52 µm. The results from compression and fatigue tests showed that the mechanical strength and durability of RHC scaffolds could be tailored by adjusting the RHC concentration, and the maximum compressive modulus reached to 0.003 MPa, which is comparable to that of BC scaffolds. The degradation test illustrated that the RHC scaffolds had a slower degradation rate compared to BC scaffolds. Finally, the biocompatibilities of the porous scaffolds were studied by seeding and culturing the human foreskin fibroblasts (HFFs) and human umbilical vein endothelial cells (HUVECs) in samples. The fluorescent images and Cell Counting Kit-8 (CCK-8) assay revealed RHC porous scaffolds were non-cytotoxic and supported the attachment as well as the proliferation of the seeded cells. Overall, the results demonstrated that RHC-based scaffolds exhibited adequate mechanical strength, ideal biodegradability, and exceptional biocompatibility, making them highly suitable for skin-tissue-engineering applications. Full article
(This article belongs to the Special Issue Biopolymers for Drug Delivery and Tissue Engineering)
Show Figures

Figure 1

14 pages, 6218 KiB  
Article
Analysis of the Effect of Human Type I Collagen-Derived Peptide on Bone Regenerative Capacity and Comparison with Various Collagen Materials In Vivo
by Tatsunori Asakura, Tran Thi Thuy Diep, Yuta Ueda, Aoi Yamada, Takahiro Tsuzuno, Naoki Takahashi, Masayuki Miyata, Koichi Tabeta, Masaki Nagata and Ken Matsuda
Medicina 2025, 61(1), 57; https://doi.org/10.3390/medicina61010057 - 2 Jan 2025
Viewed by 1498
Abstract
Background and Objectives: Autologous bone grafting is the first choice for reconstructive surgery in bone defects due to trauma or malignant tumors. However, there is an increasing demand for minimally invasive alternatives involving bone regeneration using artificial materials. Biomimetic materials that replicate [...] Read more.
Background and Objectives: Autologous bone grafting is the first choice for reconstructive surgery in bone defects due to trauma or malignant tumors. However, there is an increasing demand for minimally invasive alternatives involving bone regeneration using artificial materials. Biomimetic materials that replicate the body’s microscopic structure, such as Cellnest®, are gaining attention. Cellnest is a xeno-free recombinant peptide based on human type I collagen, containing a rich Arg-Gly-Asp (RGD) motif related to cell adhesion. The aim of this study was to compare the effects of Cellnest with existing collagen materials (Pelnac®, Integra®, Terudermis®) on bone regeneration and elucidate the underlying mechanisms. Materials and Methods: In vivo experiments involved a rat model of calvarial bone defects, in which Cellnest and other collagen materials were implanted into the defect area. Bone formation was assessed after 4 weeks using micro-computed tomography (micro-CT) and histological analysis. In vitro experiments included the 3-(4,5-dimethylthiazol-2-yl)-2,5-diphenyltetrazolium bromide (MTT), adhesion, and migration assays, and a real-time polymerase chain reaction using rapidly expanding cells (RECs) to explore the mechanisms of Cellnest’s bone regenerative capacity. Results: The micro-CT analysis showed that the regenerated bone area was significantly greater in the Cellnest group (72.3%) than in the Pelnac® (25.5%), Integra® (31.6%), and Terudermis® (38.3%) groups. The histological analysis confirmed similar trends, with Cellnest showing 42.2% bone regeneration, outperforming the other materials. The in vitro assays revealed that Cellnest promoted cell proliferation, adhesion, and migration. Gene expression analysis demonstrated that Cellnest significantly increased the levels of the bone formation markers ALP and COL1. Conclusions: Cellnest, a human type I collagen-like peptide rich in RGD motifs, enhances bone regeneration by promoting MSC adhesion and migration, and bone formation-related gene expression. The findings suggest its potential as an effective material for bone defect reconstruction. Full article
(This article belongs to the Section Dentistry and Oral Health)
Show Figures

Figure 1

15 pages, 3867 KiB  
Article
Comparison of Concentration- and Homology-Dependent Effects of the Proinflammatory Cytokine Interleukin-1β (IL-1β) in a Bovine Chondrocyte Inflammation Model
by Robert Ossendorff, Sarah Kurth, Su Wang, Max Jaenisch, Elio Assaf, Sebastian Scheidt, Kristian Welle, Christof Burger, Dieter C. Wirtz, Andreas C. Strauss and Frank A. Schildberg
Cells 2025, 14(1), 30; https://doi.org/10.3390/cells14010030 - 31 Dec 2024
Cited by 1 | Viewed by 1243
Abstract
Inflammation models with the proinflammatory cytokine interleukin-1β (IL-1β) are widely used in the in vitro investigation of new therapeutic approaches for osteoarthritis (OA). The aim of this study was to systematically analyze the influence of IL-1β in a 3D chondral pellet culture model. [...] Read more.
Inflammation models with the proinflammatory cytokine interleukin-1β (IL-1β) are widely used in the in vitro investigation of new therapeutic approaches for osteoarthritis (OA). The aim of this study was to systematically analyze the influence of IL-1β in a 3D chondral pellet culture model. Bovine articular chondrocytes were cultured to passage 3 and then placed in pellet culture. Titration of IL-1β (100–0.1 ng/mL) was performed with both human and bovine recombinant protein in chondrocyte culture for 2 weeks. Gene expression of anabolic (collagen 2, aggrecan, cartilage oligomeric protein (COMP), proteoglycan-4 (PRG-4)), catabolic matrix metallo proteinases (MMP-3, MMP-13), dedifferentiation (collagen 1) markers and inflammatory cytokines IL-6 and IL-8 was determined. Analysis of the cell culture medium was performed for the inflammatory markers IL-6 and nitric oxide (NO). In general, the influence of IL-1β was shown by a decrease in the expression of anabolic markers (collagen 2, aggrecan, PRG-4), whereas the catabolic markers MMP-3 and MMP-13 as well as the inflammatory markers IL-6 and IL-8 were significantly increased. This was observed both at the early time point (day 4) and at the late time point (day 14). The described inflammatory effects were confirmed by increased concentration-dependent release of NO and IL-6. The threshold concentration for a detectable effect compared to control differed between groups, but was reached earlier by homologous application of IL-1β. This study provides a systematic evaluation of IL-1β-specific effects on chondrocytes in a 3D pellet culture model, which is highly relevant for comparisons of studies in OA-specific drug development. Full article
Show Figures

Figure 1

15 pages, 6049 KiB  
Article
Slit1 Promotes Hypertrophic Scar Formation Through the TGF-β Signaling Pathway
by Hui Song Cui, Ya Xin Zheng, Yoon Soo Cho, Yu Mi Ro, Kibum Jeon, So Young Joo and Cheong Hoon Seo
Medicina 2024, 60(12), 2051; https://doi.org/10.3390/medicina60122051 - 12 Dec 2024
Viewed by 1610
Abstract
Background and objectives: Slit1 is a secreted protein that is closely related to cell movement and adhesion. Few studies related to fibrosis exist, and the preponderance of current research is confined to the proliferation and differentiation of neural systems. Hypertrophic scars (HTSs) are [...] Read more.
Background and objectives: Slit1 is a secreted protein that is closely related to cell movement and adhesion. Few studies related to fibrosis exist, and the preponderance of current research is confined to the proliferation and differentiation of neural systems. Hypertrophic scars (HTSs) are delineated by an overproduction of the extracellular matrix (ECM) by activated fibroblasts, leading to anomalous fibrosis, which is a severe sequela of burns. However, the functionality of Slit1 in HTS formation remains unknown. We aimed to investigate whether Slit1 regulates fibroblasts through a fibrosis-related mechanism derived from post-burn HTS tissues and normal patient tissues. Methods: Human normal fibroblasts (HNFs) and hypertrophic scar fibroblasts (HTSFs) were extracted from normal skin and post-burn HTS tissues, with settings grouped according to the patient of origin. Cell proliferation was evaluated using a CellTiter-Glo Luminescent Cell Viability Assay Kit. Cell migration experiments were carried out using a μ-Dish insert system. Protein and mRNA expression levels were quantified by Western blot and quantitative real-time polymerase chain reaction. Results: We found increased expressions of Slit1 in HTS tissues and HTSFs compared to normal tissues and HNFs. The treatment of human recombinant Slit1 protein (rSlit1) within HNFs promoted cell proliferation and differentiation, leading to an upregulation in ECM components such as α-SMA, type I and III collagen, and fibronectin. The treatment of rSlit1 in HNFs facilitated cell migration, concurrent with enhanced levels of N-cadherin and vimentin, and a diminished expression of E-cadherin. Treatment with rSlit1 resulted in the phosphorylation of SMAD pathway proteins, including SMAD2, SMAD3, and SMAD1/5/8, and non-SMAD pathway proteins, including TAK1, JNK1, ERK1/2, and p38, in HNFs. Conclusions: Exogenous Slit1 potentiates the epithelial–mesenchymal transition and upregulates SMAD and non-SMAD signaling pathways in HNFs, leading to the development of HTS, suggesting that Slit1 is a promising new target for the treatment of post-burn HTS. Full article
(This article belongs to the Special Issue Burn Injuries and Burn Rehabilitation)
Show Figures

Figure 1

20 pages, 934 KiB  
Review
What Is New in Morphea—Narrative Review on Molecular Aspects and New Targeted Therapies
by Tomasz Stein, Paulina Cieplewicz-Guźla, Katarzyna Iżykowska, Monika Pieniawska, Ryszard Żaba, Aleksandra Dańczak-Pazdrowska and Adriana Polańska
J. Clin. Med. 2024, 13(23), 7134; https://doi.org/10.3390/jcm13237134 - 25 Nov 2024
Cited by 3 | Viewed by 2914
Abstract
Morphea, also known as localized scleroderma, is an autoimmune chronic connective tissue disease. It is characterized by excessive collagen deposition in the dermis and/or subcutaneous tissue. The etiopathogenesis of this disease is not fully understood, with endothelial cell damage, immunological disorders, extracellular matrix [...] Read more.
Morphea, also known as localized scleroderma, is an autoimmune chronic connective tissue disease. It is characterized by excessive collagen deposition in the dermis and/or subcutaneous tissue. The etiopathogenesis of this disease is not fully understood, with endothelial cell damage, immunological disorders, extracellular matrix disorders and factors such as infection, trauma and other autoimmune diseases being considered. As medicine advances, there is increasing evidence that genetic factors play a significant role in disease risk and progression. In addition to environmental factors and genetic predisposition, epigenetic factors may be potential triggers for morphea. Epigenetics studies changes that affect gene expression without altering the DNA sequence, such as microRNAs, long non-coding RNAs or DNA methylation. Understanding the pathogenesis of this disease is key to identifying potential new treatments. There are anecdotal reports of good therapeutic effects following the use of biological drugs such as tocilizumab, a humanized IgG monoclonal antibody; abatacept, a recombinant soluble fusion protein; JAK inhibitors, such as tofacitinib and baricitinib; and a drug used successfully in cancer treatment, imatinib, a tyrosine kinase receptor inhibitor. In this article, we aim to review up-to-date knowledge on the pathogenesis of morphea, with particular emphasis on genetic and epigenetic factors. In addition, we present the new options of morphea treatment based on several case series treated with new drugs that are potential targets for the development of therapies for this disease. Full article
(This article belongs to the Section Immunology)
Show Figures

Figure 1

28 pages, 13740 KiB  
Article
A Novel P-III Metalloproteinase from Bothrops barnetti Venom Degrades Extracellular Matrix Proteins, Inhibits Platelet Aggregation, and Disrupts Endothelial Cell Adhesion via α5β1 Integrin Receptors to Arginine–Glycine–Aspartic Acid (RGD)-Containing Molecules
by Pedro Henrique de Caires Schluga, Debora Larangote, Ana Maria de Melo, Guilherme Kamienski Lobermayer, Daniel Torrejón, Luciana Souza de Oliveira, Valeria Gonçalves Alvarenga, Dan Erick Vivas-Ruiz, Silvio Sanches Veiga, Eladio Flores Sanchez and Luiza Helena Gremski
Toxins 2024, 16(11), 486; https://doi.org/10.3390/toxins16110486 - 9 Nov 2024
Cited by 2 | Viewed by 2097
Abstract
Viperid snake venoms are notably abundant in metalloproteinases (proteins) (SVMPs), which are primarily responsible for inducing hemorrhage and disrupting the hemostatic process and tissue integrity in envenomed victims. In this study, barnettlysin-III (Bar-III), a hemorrhagic P-III SVMP, was purified from the venom of [...] Read more.
Viperid snake venoms are notably abundant in metalloproteinases (proteins) (SVMPs), which are primarily responsible for inducing hemorrhage and disrupting the hemostatic process and tissue integrity in envenomed victims. In this study, barnettlysin-III (Bar-III), a hemorrhagic P-III SVMP, was purified from the venom of the Peruvian snake Bothrops barnetti. Bar-III has a molecular mass of approximately 50 kDa and is a glycosylation-dependent functional metalloproteinase. Some biochemical properties of Bar-III, including the full amino acid sequence deduced from its cDNA, are reported. Its enzymatic activity is increased by Ca2+ ions and inhibited by an excess of Zn2+. Synthetic metalloproteinase inhibitors and EDTA also inhibit its proteolytic action. Bar-III degrades several plasma and ECM proteins, including fibrin(ogen), fibronectin, laminin, and nidogen. Platelets play a key role in hemostasis and thrombosis and in other biological process, such as inflammation and immunity, and platelet activation is driven by the platelet signaling receptors, glycoprotein (GP)Ib-IX-V, which binds vWF, and GPVI, which binds collagen. Moreover, Bar-III inhibits vWF- and convulxin-induced platelet aggregation in human washed platelets by cleaving the recombinant A1 domain of vWF and GPVI into a soluble ectodomain fraction of ~55 kDa (sGPVI). Bar-III does not reduce the viability of cultured endothelial cells; however, it interferes with the adhesion of these cells to fibronectin, vitronectin, and RGD peptides, as well as their migration profile. Bar-III binds specifically to the surface of these cells, and part of this interaction involves α5β1 integrin receptors. These results contribute to a better comprehension of the pathophysiology of snakebite accidents/incidents and could be used as a tool to explore novel and safer anti-venom therapeutics. Full article
(This article belongs to the Section Animal Venoms)
Show Figures

Figure 1

13 pages, 2514 KiB  
Article
IL-11 Expression in Systemic Sclerosis Is Dependent on Caspase-1 Activity but Does Not Increase Collagen Deposition
by Caya M. McFalls, Lianne M. Connolly, Alfred G. Fustakgi and Carol M. Artlett
Rheumato 2024, 4(4), 163-175; https://doi.org/10.3390/rheumato4040013 - 12 Oct 2024
Cited by 1 | Viewed by 1435
Abstract
Background: Interleukin-11 (IL-11) is increased in patients with systemic sclerosis (SSc) and is thought to play a role in fibrosis. Many studies have reported decreased fibrosis when IL-11 is blocked, but few have examined factors that induce IL-11 expression. Because fibrosis has been [...] Read more.
Background: Interleukin-11 (IL-11) is increased in patients with systemic sclerosis (SSc) and is thought to play a role in fibrosis. Many studies have reported decreased fibrosis when IL-11 is blocked, but few have examined factors that induce IL-11 expression. Because fibrosis has been linked to activated inflammasomes driving caspase-1 maturation and the secretion of IL-1β, we set out to determine if IL-11 expression was dependent on caspase-1 activity. Methods: Primary lung fibroblast cell lines derived from patients with SSc, IPF (fibrotic control), and healthy individuals were cultured at low passage. Gene expression for IL-11 and the IL-11 receptor (IL-11Rα1) was analyzed using qPCR and normalized to the control, and collagen production was measured using Sirius Red. Results: SSc and IPF fibroblasts expressed significantly more IL-11 transcripts than normal cells (3.35-fold and 9.97-fold more, p = 0.0396 and p = 0.0023, respectively). IL-11Rα1 was expressed 2.32-fold and 2.27-fold more in SSc and IPF (p = 0.0004 and p = 0.0032, respectively) than in normal cells. In SSc fibroblasts, inhibition of caspase-1 with YVAD decreased IL-11 expression by 49.59% (p = 0.0016) but did not affect IL-11Rα1 expression (p > 0.05). IL-11 expression was increased 2.97-fold with TGF-β1 (p = 0.0030) and 22.24-fold with IL-1β (p < 0.0001), while the expression of IL-11Rα1 was not induced with these two cytokines. LPS increased IL-11 expression in normal fibroblasts 1.52-fold (p = 0.0042), which was abolished with YVAD (p < 0.0001). IL-11Rα1 gene transcripts were also increased with LPS 1.50-fold (p = 0.0132), but YVAD did not inhibit this expression. In these studies, we were unable to detect IL-11 protein nor were we able to induce COL1A1 expression or increase the total amount of collagen secreted by fibroblasts with human recombinant IL-11. Conclusions: IL-11 and its receptor, IL-11Rα1, are both elevated in fibrosis. IL-11 expression is dependent on inflammasome activation of caspase-1 and the downstream cytokines TGF-β1 and IL-1β, while IL-11Rα1 was only dependent on NF-kB. Full article
Show Figures

Figure 1

17 pages, 2880 KiB  
Article
Preparation and Characterization of Hydroxylated Recombinant Collagen by Incorporating Proline and Hydroxyproline in Proline-Deficient Escherichia coli
by Zhimin Cheng, Bin Hong, Yanmei Li and Jufang Wang
Bioengineering 2024, 11(10), 975; https://doi.org/10.3390/bioengineering11100975 - 27 Sep 2024
Cited by 1 | Viewed by 2446
Abstract
Collagen possesses distinctive chemical properties and biological functions due to its unique triple helix structure. However, recombinant collagen expressed in Escherichia coli without post-translational modifications such as hydroxylation lacks full function since hydroxylation is considered to be critical to the stability of the [...] Read more.
Collagen possesses distinctive chemical properties and biological functions due to its unique triple helix structure. However, recombinant collagen expressed in Escherichia coli without post-translational modifications such as hydroxylation lacks full function since hydroxylation is considered to be critical to the stability of the collagen triple-helix at body temperature. Here, a proline-deficient E. coli strain was constructed and employed to prepare hydroxylated recombinant collagens by incorporating proline (Pro) and hydroxyproline (Hyp) from the culture medium. By controlling the ratio of Pro to Hyp in the culture medium, collagen with different degrees of hydroxylation (0–88%) can be obtained. When the ratio of Pro and Hyp was adjusted to 12:8 mM, the proline hydroxylation rate of recombinant human collagen (rhCol, 55 kDa) ranged from 40–50%, which was also the degree of natural collagen. After proline hydroxylation, both the thermal stability and cell binding of rhCol were significantly enhanced. Notably, when the hydroxylation rate approached that of native human collagen (40–50%), the improvements were most pronounced. Moreover, the cell binding of rhCol with a hydroxylation rate of 43% increased by 29%, and the melting temperature (Tm) rose by 5 °C compared to the non-hydroxylated rhCol. The system achieved a yield of 1.186 g/L of rhCol by batch-fed in a 7 L fermenter. This innovative technology is expected to drive the development and application of collagen-related biomaterials with significant application value in the fields of tissue engineering, regenerative medicine, and biopharmaceuticals. Full article
(This article belongs to the Special Issue Bio-Applications of Engineered Escherichia coli)
Show Figures

Figure 1

Back to TopTop