251
|
Zhu Y, Wang L, Liu R, Ding X, Yin S, Chen Y, Zhu C, Wang Z, Li W. Inhibition of PRMT1 alleviates sepsis-induced acute kidney injury in mice by blocking the TGF-β1 and IL-6 trans-signaling pathways. FEBS Open Bio 2023; 13:1859-1873. [PMID: 37525933 PMCID: PMC10549220 DOI: 10.1002/2211-5463.13684] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2022] [Revised: 06/28/2023] [Accepted: 07/31/2023] [Indexed: 08/02/2023] Open
Abstract
Sepsis-induced acute kidney injury (SI-AKI) causes renal dysfunction and has a high mortality rate. Protein arginine methyltransferase-1 (PRMT1) is a key regulator of renal insufficiency. In the present study, we explored the potential involvement of PRMT1 in SI-AKI. A murine model of SI-AKI was induced by cecal ligation and perforation. The expression and localization of PRMT1 and molecules involved in the transforming growth factor (TGF)-β1/Smad3 and interleukin (IL)-6/signal transducer and activator of transcription 3 (STAT3) signaling pathways were detected in mouse kidney tissues by western blot analysis, immunofluorescence, and immunohistochemistry. The association of PRMT1 with downstream molecules of the TGF-β1/Smad3 and IL-6/STAT3 signaling pathways was further verified in vitro in mouse renal tubular epithelial cells. Cecal ligation and perforation caused epithelial-mesenchymal transition, apoptosis, and inflammation in renal tissues, and this was alleviated by inhibition of PRMT1. Inhibition of PRMT1 in SI-AKI mice decreased the expression of TGF-β1 and phosphorylation of Smad3 in the renal cortex, and downregulated the expression of soluble IL-6R and phosphorylation of STAT3 in the medulla. Knockdown of PRMT1 in mouse renal tubular epithelial cells restricted the expression of Cox-2, E-cadherin, Pro-caspase3, and phosphorylated Smad3 (involved in the TGF-β1-mediated signaling pathway), and also blocked IL-6/soluble IL-6R, inducing the expression of Cox-2 and phosphorylated-STAT3. In conclusion, our findings suggest that inhibition of PRMT1 mitigates SI-AKI by inactivating the TGF-β1/Smad3 pathway in the cortex and the IL-6/STAT3 pathway in the medulla. Our findings may aid in the identification of potential therapeutic target molecules for SI-AKI.
Collapse
Affiliation(s)
- Yu Zhu
- Nephrology Department, Shenzhen HospitalUniversity of Chinese Academy of Sciences (Guangming)ShenzhenChina
| | - Longmei Wang
- Department of Infectious DiseasesEnze Medical CenterLinhaiChina
| | - Rui Liu
- Department of Infectious and Tropical DiseasesThe Second Affiliated Hospital of Hainan Medical UniversityHaikouChina
- National Health Commission Key Laboratory of Tropical Disease ControlHainan Medical UniversityHaikouChina
| | | | - Song Yin
- Division of Life Sciences and Medicine, Department of Infectious Disease, The First Affiliated Hospital of USTCUniversity of Science and Technology of ChinaHefeiChina
- Wannan Medical CollegeWuhuChina
| | - Yuankun Chen
- Department of Infectious and Tropical DiseasesThe Second Affiliated Hospital of Hainan Medical UniversityHaikouChina
- National Health Commission Key Laboratory of Tropical Disease ControlHainan Medical UniversityHaikouChina
| | - Chuanlong Zhu
- Department of Infectious and Tropical DiseasesThe Second Affiliated Hospital of Hainan Medical UniversityHaikouChina
- Department of Infectious DiseaseThe First Affiliated Hospital of Nanjing Medical UniversityChina
| | - Zheng Wang
- Department of Respiratory and Critical MedicinePeople's Hospital of Zhengzhou UniversityChina
| | - Wenting Li
- Department of Infectious and Tropical DiseasesThe Second Affiliated Hospital of Hainan Medical UniversityHaikouChina
- National Health Commission Key Laboratory of Tropical Disease ControlHainan Medical UniversityHaikouChina
- Department of Infectious DiseaseThe First Affiliated Hospital of Anhui Medical UniversityHefeiChina
| |
Collapse
|
252
|
Yang Y, Chu C, Liu L, Wang C, Hu C, Rung S, Man Y, Qu Y. Tracing immune cells around biomaterials with spatial anchors during large-scale wound regeneration. Nat Commun 2023; 14:5995. [PMID: 37752124 PMCID: PMC10522601 DOI: 10.1038/s41467-023-41608-9] [Citation(s) in RCA: 17] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2022] [Accepted: 09/11/2023] [Indexed: 09/28/2023] Open
Abstract
Skin scarring devoid of dermal appendages after severe trauma has unfavorable effects on aesthetic and physiological functions. Here we present a method for large-area wound regeneration using biodegradable aligned extracellular matrix scaffolds. We show that the implantation of these scaffolds accelerates wound coverage and enhances hair follicle neogenesis. We perform multimodal analysis, in combination with single-cell RNA sequencing and spatial transcriptomics, to explore the immune responses around biomaterials, highlighting the potential role of regulatory T cells in mitigating tissue fibrous by suppressing excessive type 2 inflammation. We find that immunodeficient mice lacking mature T lymphocytes show the typical characteristic of tissue fibrous driven by type 2 macrophage inflammation, validating the potential therapeutic effect of the adaptive immune system activated by biomaterials. These findings contribute to our understanding of the coordination of immune systems in wound regeneration and facilitate the design of immunoregulatory biomaterials in the future.
Collapse
Affiliation(s)
- Yang Yang
- Department of Oral Implantology & State Key Laboratory of Oral Diseases and National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, 610041, China
- Department of Prosthodontics & State Key Laboratory of Oral Diseases and National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, 610041, China
| | - Chenyu Chu
- Department of Oral Implantology & State Key Laboratory of Oral Diseases and National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, 610041, China
| | - Li Liu
- Department of Oral Implantology & State Key Laboratory of Oral Diseases and National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, 610041, China
| | - Chenbing Wang
- Department of Oral Implantology & State Key Laboratory of Oral Diseases and National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, 610041, China
| | - Chen Hu
- Department of Oral Implantology & State Key Laboratory of Oral Diseases and National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, 610041, China
| | - Shengan Rung
- Department of Oral Implantology & State Key Laboratory of Oral Diseases and National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, 610041, China
- Department of Prosthodontics & State Key Laboratory of Oral Diseases and National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, 610041, China
| | - Yi Man
- Department of Oral Implantology & State Key Laboratory of Oral Diseases and National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, 610041, China.
| | - Yili Qu
- Department of Prosthodontics & State Key Laboratory of Oral Diseases and National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, 610041, China.
| |
Collapse
|
253
|
Yilmaz K, Haeberle S, Kim YO, Fritzler MJ, Weng SY, Goeppert B, Raker VK, Steinbrink K, Schuppan D, Enk A, Hadaschik EN. Regulatory T-cell deficiency leads to features of autoimmune liver disease overlap syndrome in scurfy mice. Front Immunol 2023; 14:1253649. [PMID: 37818371 PMCID: PMC10561387 DOI: 10.3389/fimmu.2023.1253649] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2023] [Accepted: 09/08/2023] [Indexed: 10/12/2023] Open
Abstract
Introduction Scurfy mice have a complete deficiency of functional regulatory T cells (Treg) due to a frameshift mutation in the Foxp3 gene. The impaired immune homeostasis results in a lethal lymphoproliferative disorder affecting multiple organs, including the liver. The autoimmune pathology in scurfy mice is in part accompanied by autoantibodies such as antinuclear antibodies (ANA). ANA are serological hallmarks of several autoimmune disorders including autoimmune liver diseases (AILD). However, the underlying pathogenesis and the role of Treg in AILD remain to be elucidated. The present study therefore aimed to characterize the liver disease in scurfy mice. Methods Sera from scurfy mice were screened for ANA by indirect immunofluorescence assay (IFA) and tested for a wide range of AILD-associated autoantibodies by enzyme-linked immunosorbent assay, line immunoassay, and addressable laser bead immunoassay. CD4+ T cells of scurfy mice were transferred into T cell-deficient B6/nude mice. Monoclonal autoantibodies from scurfy mice and recipient B6/nude mice were tested for ANA by IFA. Liver tissue of scurfy mice was analyzed by conventional histology. Collagen deposition in scurfy liver was quantified via hepatic hydroxyproline content. Real-time quantitative PCR was used to determine fibrosis-related hepatic gene expression. Hepatic immune cells were differentiated by flow cytometry. Results All scurfy mice produced ANA. AILD-associated autoantibodies, predominantly antimitochondrial antibodies, were detected at significantly higher levels in scurfy sera. CD4+ T cells from scurfy mice were sufficient to induce anti-dsDNA autoantibodies and ANA with an AILD-related nuclear envelope staining pattern. Liver histology revealed portal inflammation with bile duct damage and proliferation, as in primary biliary cholangitis (PBC), and interface hepatitis with portal-parenchymal necroinflammation, as found in autoimmune hepatitis (AIH). In scurfy liver, TNFα and fibrosis-related transcripts including Col1a1, Timp1, Acta2, Mmp2, and Mmp9 were upregulated. The level of proinflammatory monocytic macrophages (Ly-6Chi) was increased, while M2-type macrophages (CD206+) were downregulated compared to wildtype controls. Despite severe hepatic inflammation, fibrosis did not develop within 25 days, which is close to the lifespan of scurfy mice. Discussion Our findings suggest that Treg-deficient scurfy mice spontaneously develop clinical, serological, and immunopathological characteristics of AILD with overlapping features of PBC and AIH.
Collapse
Affiliation(s)
- Kaan Yilmaz
- Department of Dermatology, University of Heidelberg, Heidelberg, Germany
- Department of Dermatology, University Medical Center Mannheim, Medical Faculty Mannheim, University of Heidelberg, Mannheim, Germany
| | - Stefanie Haeberle
- Department of Dermatology, University of Heidelberg, Heidelberg, Germany
| | - Yong Ook Kim
- Institute of Translational Immunology, University Medical Center of Johannes Gutenberg University Mainz, Mainz, Germany
| | - Marvin J. Fritzler
- Department of Medicine, Cumming School of Medicine, University of Calgary, Calgary, AB, Canada
| | - Shih-Yen Weng
- Institute of Translational Immunology, University Medical Center of Johannes Gutenberg University Mainz, Mainz, Germany
- Smart Healthcare Interdisciplinary College, National Taipei University of Nursing and Health Sciences, Taipei, Taiwan
| | - Benjamin Goeppert
- Institute of Tissue Medicine and Pathology, University of Bern, Bern, Switzerland
- Institute of Pathology and Neuropathology, RKH Klinikum Ludwigsburg, Ludwigsburg, Germany
| | - Verena K. Raker
- Department of Dermatology, University Hospital Muenster, Muenster, Germany
| | - Kerstin Steinbrink
- Department of Dermatology, University Hospital Muenster, Muenster, Germany
| | - Detlef Schuppan
- Institute of Translational Immunology, University Medical Center of Johannes Gutenberg University Mainz, Mainz, Germany
- Division of Gastroenterology, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA, United States
| | - Alexander Enk
- Department of Dermatology, University of Heidelberg, Heidelberg, Germany
| | - Eva N. Hadaschik
- Department of Dermatology, University of Heidelberg, Heidelberg, Germany
- Department of Dermatology, University Hospital of Essen, Essen, Germany
| |
Collapse
|
254
|
Kumar V, Hertz M, Agro A, Byrne AJ. Type 1 invariant natural killer T cells in chronic inflammation and tissue fibrosis. Front Immunol 2023; 14:1260503. [PMID: 37818376 PMCID: PMC10561218 DOI: 10.3389/fimmu.2023.1260503] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2023] [Accepted: 09/06/2023] [Indexed: 10/12/2023] Open
Abstract
Chronic tissue inflammation often results in fibrosis characterized by the accumulation of extracellular matrix components remodeling normal tissue architecture and function. Recent studies have suggested common immune mechanisms despite the complexity of the interactions between tissue-specific fibroblasts, macrophages, and distinct immune cell populations that mediate fibrosis in various tissues. Natural killer T (NKT) cells recognizing lipid antigens bound to CD1d molecules have been shown to play an important role in chronic inflammation and fibrosis. Here we review recent data in both experimental models and in humans that suggest a key role of type 1 invariant NKT (iNKT) cell activation in the progression of inflammatory cascades leading to recruitment of neutrophils and activation of the inflammasome, macrophages, fibroblasts, and, ultimately, fibrosis. Emerging evidence suggests that iNKT-associated mechanisms contribute to type 1, type 2 and type 3 immune pathways mediating tissue fibrosis, including idiopathic pulmonary fibrosis (IPF). Thus, targeting a pathway upstream of these immune mechanisms, such as the inhibition of iNKT activation, may be important in modulating various fibrotic conditions.
Collapse
Affiliation(s)
- Vipin Kumar
- Laboratory of Immune Regulation, Department of Medicine, University of California San Diego, La Jolla, CA, United States
- GRI Bio, La Jolla, CA, United States
| | | | | | - Adam J. Byrne
- National Heart and Lung Institute, Imperial College London, London, United Kingdom
- School of Medicine and Conway Institute for Biomolecular and Biomedical Research, University College Dublin, Dublin, Ireland
| |
Collapse
|
255
|
Jiang L, Liu T, Lyu K, Chen Y, Lu J, Wang X, Long L, Li S. Inflammation-related signaling pathways in tendinopathy. Open Life Sci 2023; 18:20220729. [PMID: 37744452 PMCID: PMC10512452 DOI: 10.1515/biol-2022-0729] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2023] [Revised: 08/23/2023] [Accepted: 08/24/2023] [Indexed: 09/26/2023] Open
Abstract
Tendon is a connective tissue that produces movement by transmitting the force produced by muscle contraction to the bones. Most tendinopathy is caused by prolonged overloading of the tendon, leading to degenerative disease of the tendon. When overloaded, the oxygen demand of tenocytes increases, and the tendon structure is special and lacks blood supply, which makes it easier to form an oxygen-deficient environment in tenocytes. The production of reactive oxygen species due to hypoxia causes elevation of inflammatory markers in the tendon, including PGE2, IL-1β, and TNF-α. In the process of tendon healing, inflammation is also a necessary stage. The inflammatory environment formed by cytokines and various immune cells play an important role in the clearance of necrotic material, the proliferation of tenocytes, and the production of collagen fibers. However, excessive inflammation can lead to tendon adhesions and hinder tendon healing. Some important and diverse biological functions of the body originate from intercellular signal transduction, among which cytokine mediation is an important way of signal transduction. In particular, NF-κB, NLRP3, p38/MAPK, and signal transducer and activator of transcription 3, four common signaling pathways in tendinopathy inflammatory response, play a crucial role in the regulation and transcription of inflammatory factors. Therefore, summarizing the specific mechanisms of inflammatory signaling pathways in tendinopathy is of great significance for an in-depth understanding of the inflammatory response process and exploring how to inhibit the harmful part of the inflammatory response and promote the beneficial part to improve the healing effect of the tendon.
Collapse
Affiliation(s)
- Li Jiang
- School of Physical Education, Southwest Medical University, Luzhou, 646000, China
| | - Tianzhu Liu
- Neurology Department, The Affiliated Traditional Chinese Medicine Hospital of Southwest Medical University, Luzhou, 646000, China
| | - Kexin Lyu
- School of Physical Education, Southwest Medical University, Luzhou, 646000, China
| | - Yixuan Chen
- School of Physical Education, Southwest Medical University, Luzhou, 646000, China
| | - Jingwei Lu
- School of Physical Education, Southwest Medical University, Luzhou, 646000, China
| | - Xiaoqiang Wang
- The Affiliated Traditional Chinese Medicine Hospital of Southwest Medical University, Luzhou, 646000, China
| | - Longhai Long
- The Affiliated Traditional Chinese Medicine Hospital of Southwest Medical University, Luzhou, 646000, China
| | - Sen Li
- Division of Spine Surgery, Department of Orthopedic Surgery, Nanjing Drum Tower Hospital, Affiliated Hospital
of Medical School, Nanjing University, Nanjing, 210000, China
| |
Collapse
|
256
|
Mishchenko O, Yanovska A, Kosinov O, Maksymov D, Moskalenko R, Ramanavicius A, Pogorielov M. Synthetic Calcium-Phosphate Materials for Bone Grafting. Polymers (Basel) 2023; 15:3822. [PMID: 37765676 PMCID: PMC10536599 DOI: 10.3390/polym15183822] [Citation(s) in RCA: 21] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2023] [Revised: 09/08/2023] [Accepted: 09/11/2023] [Indexed: 09/29/2023] Open
Abstract
Synthetic bone grafting materials play a significant role in various medical applications involving bone regeneration and repair. Their ability to mimic the properties of natural bone and promote the healing process has contributed to their growing relevance. While calcium-phosphates and their composites with various polymers and biopolymers are widely used in clinical and experimental research, the diverse range of available polymer-based materials poses challenges in selecting the most suitable grafts for successful bone repair. This review aims to address the fundamental issues of bone biology and regeneration while providing a clear perspective on the principles guiding the development of synthetic materials. In this study, we delve into the basic principles underlying the creation of synthetic bone composites and explore the mechanisms of formation for biologically important complexes and structures associated with the various constituent parts of these materials. Additionally, we offer comprehensive information on the application of biologically active substances to enhance the properties and bioactivity of synthetic bone grafting materials. By presenting these insights, our review enables a deeper understanding of the regeneration processes facilitated by the application of synthetic bone composites.
Collapse
Affiliation(s)
- Oleg Mishchenko
- Department of Surgical and Propaedeutic Dentistry, Zaporizhzhia State Medical and Pharmaceutical University, 26, Prosp. Mayakovskogo, 69035 Zaporizhzhia, Ukraine; (O.M.); (O.K.); (D.M.)
| | - Anna Yanovska
- Theoretical and Applied Chemistry Department, Sumy State University, R-Korsakova Street, 40007 Sumy, Ukraine
| | - Oleksii Kosinov
- Department of Surgical and Propaedeutic Dentistry, Zaporizhzhia State Medical and Pharmaceutical University, 26, Prosp. Mayakovskogo, 69035 Zaporizhzhia, Ukraine; (O.M.); (O.K.); (D.M.)
| | - Denys Maksymov
- Department of Surgical and Propaedeutic Dentistry, Zaporizhzhia State Medical and Pharmaceutical University, 26, Prosp. Mayakovskogo, 69035 Zaporizhzhia, Ukraine; (O.M.); (O.K.); (D.M.)
| | - Roman Moskalenko
- Department of Pathology, Sumy State University, R-Korsakova Street, 40007 Sumy, Ukraine;
| | - Arunas Ramanavicius
- NanoTechnas-Center of Nanotechnology and Materials Science, Institute of Chemistry, Faculty of Chemistry and Geosciences, Vilnius University, Naugarduko Str. 24, LT-03225 Vilnius, Lithuania
| | - Maksym Pogorielov
- Biomedical Research Centre, Sumy State University, R-Korsakova Street, 40007 Sumy, Ukraine;
- Institute of Atomic Physics and Spectroscopy, University of Latvia, Jelgavas Iela 3, LV-1004 Riga, Latvia
| |
Collapse
|
257
|
Zangiabadi S, Chamoun KP, Nguyen K, Tang Y, Sweeney G, Abdul-Sater AA. Copper infused fabric attenuates inflammation in macrophages. PLoS One 2023; 18:e0287741. [PMID: 37713400 PMCID: PMC10503751 DOI: 10.1371/journal.pone.0287741] [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: 01/05/2023] [Accepted: 06/12/2023] [Indexed: 09/17/2023] Open
Abstract
While inflammation is an important immune response for protection from infections, excessive or prolonged inflammation can lead to a variety of debilitating diseases including skin disease, diabetes, heart disease, stroke, autoimmune diseases and cancer. Inflammation is a graded response that is typically initiated when resident macrophages sense the presence of pathogens or damage in the tissue and produce inflammatory cytokines and chemokines to kill the pathogen, clear debris and dead tissue, and initiate tissue repair. Here we show that copper-infused fabrics can prevent inflammation by blocking the production of inflammatory cytokines from macrophages after being exposed to LPS, a component of bacterial cell wall. Mechanistically, we show that copper-infused fabrics can significantly reduce the NF-κB and IRF3 activation in LPS-stimulated macrophages. Given the importance of excessive inflammation in diabetes, we show that copper can reduce insulin resistance mediated by inflammatory cytokines in muscle cells. Our data show that copper infused fabrics may be useful to reduce excessive inflammation in macrophages and improve insulin sensitivity in skeletal muscles.
Collapse
Affiliation(s)
- Safoura Zangiabadi
- School of Kinesiology and Health Science, Muscle Health Research Centre (MHRC), York University, Toronto, Canada
| | - Khalil P. Chamoun
- School of Kinesiology and Health Science, Muscle Health Research Centre (MHRC), York University, Toronto, Canada
| | - Khang Nguyen
- Department of Biology, York University, Toronto, Ontario, Canada
| | - Yitian Tang
- School of Kinesiology and Health Science, Muscle Health Research Centre (MHRC), York University, Toronto, Canada
| | - Gary Sweeney
- Department of Biology, York University, Toronto, Ontario, Canada
| | - Ali A. Abdul-Sater
- School of Kinesiology and Health Science, Muscle Health Research Centre (MHRC), York University, Toronto, Canada
| |
Collapse
|
258
|
Hunt M, Torres M, Bachar-Wikström E, Wikström JD. Multifaceted roles of mitochondria in wound healing and chronic wound pathogenesis. Front Cell Dev Biol 2023; 11:1252318. [PMID: 37771375 PMCID: PMC10523588 DOI: 10.3389/fcell.2023.1252318] [Citation(s) in RCA: 15] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2023] [Accepted: 08/28/2023] [Indexed: 09/30/2023] Open
Abstract
Mitochondria are intracellular organelles that play a critical role in numerous cellular processes including the regulation of metabolism, cellular stress response, and cell fate. Mitochondria themselves are subject to well-orchestrated regulation in order to maintain organelle and cellular homeostasis. Wound healing is a multifactorial process that involves the stringent regulation of several cell types and cellular processes. In the event of dysregulated wound healing, hard-to-heal chronic wounds form and can place a significant burden on healthcare systems. Importantly, treatment options remain limited owing to the multifactorial nature of chronic wound pathogenesis. One area that has received more attention in recent years is the role of mitochondria in wound healing. With regards to this, current literature has demonstrated an important role for mitochondria in several areas of wound healing and chronic wound pathogenesis including metabolism, apoptosis, and redox signalling. Additionally, the influence of mitochondrial dynamics and mitophagy has also been investigated. However, few studies have utilised patient tissue when studying mitochondria in wound healing, instead using various animal models. In this review we dissect the current knowledge of the role of mitochondria in wound healing and discuss how future research can potentially aid in the progression of wound healing research.
Collapse
Affiliation(s)
- Matthew Hunt
- Dermatology and Venerology Division, Department of Medicine (Solna), Karolinska Institutet, Stockholm, Sweden
| | - Monica Torres
- Dermatology and Venerology Division, Department of Medicine (Solna), Karolinska Institutet, Stockholm, Sweden
- Dermato-Venereology Clinic, Karolinska University Hospital, Stockholm, Sweden
| | - Etty Bachar-Wikström
- Dermatology and Venerology Division, Department of Medicine (Solna), Karolinska Institutet, Stockholm, Sweden
| | - Jakob D. Wikström
- Dermatology and Venerology Division, Department of Medicine (Solna), Karolinska Institutet, Stockholm, Sweden
- Dermato-Venereology Clinic, Karolinska University Hospital, Stockholm, Sweden
| |
Collapse
|
259
|
Qiu B, Lawan A, Xirouchaki CE, Yi JS, Robert M, Zhang L, Brown W, Fernández-Hernando C, Yang X, Tiganis T, Bennett AM. MKP1 promotes nonalcoholic steatohepatitis by suppressing AMPK activity through LKB1 nuclear retention. Nat Commun 2023; 14:5405. [PMID: 37669951 PMCID: PMC10480499 DOI: 10.1038/s41467-023-41145-5] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2023] [Accepted: 08/24/2023] [Indexed: 09/07/2023] Open
Abstract
Nonalcoholic steatohepatitis (NASH) is triggered by hepatocyte death through activation of caspase 6, as a result of decreased adenosine monophosphate (AMP)-activated protein kinase-alpha (AMPKα) activity. Increased hepatocellular death promotes inflammation which drives hepatic fibrosis. We show that the nuclear-localized mitogen-activated protein kinase (MAPK) phosphatase-1 (MKP1) is upregulated in NASH patients and in NASH diet fed male mice. The focus of this work is to investigate whether and how MKP1 is involved in the development of NASH. Under NASH conditions increased oxidative stress, induces MKP1 expression leading to nuclear p38 MAPK dephosphorylation and decreases liver kinase B1 (LKB1) phosphorylation at a site required to promote LKB1 nuclear exit. Hepatic deletion of MKP1 in NASH diet fed male mice releases nuclear LKB1 into the cytoplasm to activate AMPKα and prevents hepatocellular death, inflammation and NASH. Hence, nuclear-localized MKP1-p38 MAPK-LKB1 signaling is required to suppress AMPKα which triggers hepatocyte death and the development of NASH.
Collapse
Affiliation(s)
- Bin Qiu
- Yale University School of Medicine, Department of Pharmacology, 333 Cedar Street, New Haven, CT, 06520, USA
- Yale University School of Medicine, Yale Center of Molecular and Systems Metabolism, New Haven, CT, 06520, USA
| | - Ahmed Lawan
- University of Alabama, Department of Biological Sciences, 301 Sparkman Drive, Huntsville, AL, 35899, USA
| | - Chrysovalantou E Xirouchaki
- Monash Biomedicine Discovery Institute, Monash University, Clayton, VIC, 3800, Australia
- Department of Biochemistry and Molecular Biology, Monash University, Clayton, VIC, 3800, Australia
| | - Jae-Sung Yi
- Yale University School of Medicine, Department of Pharmacology, 333 Cedar Street, New Haven, CT, 06520, USA
- Yale University School of Medicine, Yale Center of Molecular and Systems Metabolism, New Haven, CT, 06520, USA
| | - Marie Robert
- Yale University School of Medicine, Department of Pathology, 300 Cedar Street, New Haven, CT, 06520, USA
| | - Lei Zhang
- Yale University School of Medicine, Department of Pharmacology, 333 Cedar Street, New Haven, CT, 06520, USA
- Yale University School of Medicine, Yale Center of Molecular and Systems Metabolism, New Haven, CT, 06520, USA
| | - Wendy Brown
- Monash University Department of Surgery, Alfred Hospital, Melbourne, Victoria, 3004, Australia
| | - Carlos Fernández-Hernando
- Yale University School of Medicine, Yale Center of Molecular and Systems Metabolism, New Haven, CT, 06520, USA
- Yale University School of Medicine, Department of Pathology, 300 Cedar Street, New Haven, CT, 06520, USA
- Yale University School of Medicine, Vascular Biology and Therapeutics Program, New Haven, CT, 06520, USA
- Department of Comparative Medicine, Yale University School of Medicine, New Haven, CT, USA
| | - Xiaoyong Yang
- Yale University School of Medicine, Yale Center of Molecular and Systems Metabolism, New Haven, CT, 06520, USA
- Department of Comparative Medicine, Yale University School of Medicine, New Haven, CT, USA
| | - Tony Tiganis
- Monash Biomedicine Discovery Institute, Monash University, Clayton, VIC, 3800, Australia
- Department of Biochemistry and Molecular Biology, Monash University, Clayton, VIC, 3800, Australia
- Department of Comparative Medicine, Yale University School of Medicine, New Haven, CT, USA
| | - Anton M Bennett
- Yale University School of Medicine, Department of Pharmacology, 333 Cedar Street, New Haven, CT, 06520, USA.
- Yale University School of Medicine, Yale Center of Molecular and Systems Metabolism, New Haven, CT, 06520, USA.
- Yale University School of Medicine, Vascular Biology and Therapeutics Program, New Haven, CT, 06520, USA.
- Department of Comparative Medicine, Yale University School of Medicine, New Haven, CT, USA.
| |
Collapse
|
260
|
Han H, Xing L, Chen BT, Liu Y, Zhou TJ, Wang Y, Zhang LF, Li L, Cho CS, Jiang HL. Progress on the pathological tissue microenvironment barrier-modulated nanomedicine. Adv Drug Deliv Rev 2023; 200:115051. [PMID: 37549848 DOI: 10.1016/j.addr.2023.115051] [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: 06/18/2023] [Revised: 07/21/2023] [Accepted: 08/04/2023] [Indexed: 08/09/2023]
Abstract
Imbalance in the tissue microenvironment is the main obstacle to drug delivery and distribution in the human body. Before penetrating the pathological tissue microenvironment to the target site, therapeutic agents are usually accompanied by three consumption steps: the first step is tissue physical barriers for prevention of their penetration, the second step is inactivation of them by biological molecules, and the third step is a cytoprotective mechanism for preventing them from functioning on specific subcellular organelles. However, recent studies in drug-hindering mainly focus on normal physiological rather than pathological microenvironment, and the repair of damaged physiological barriers is also rarely discussed. Actually, both the modulation of pathological barriers and the repair of damaged physiological barriers are essential in the disease treatment and the homeostasis maintenance. In this review, we present an overview describing the latest advances in the generality of these pathological barriers and barrier-modulated nanomedicine. Overall, this review holds considerable significance for guiding the design of nanomedicine to increase drug efficacy in the future.
Collapse
Affiliation(s)
- Han Han
- State Key Laboratory of Natural Medicines, China Pharmaceutical University, Nanjing 211198, China
| | - Lei Xing
- State Key Laboratory of Natural Medicines, China Pharmaceutical University, Nanjing 211198, China; College of Pharmacy, Yanbian University, Yanji 133002, China
| | - Bi-Te Chen
- State Key Laboratory of Natural Medicines, China Pharmaceutical University, Nanjing 211198, China
| | - Yang Liu
- State Key Laboratory of Natural Medicines, China Pharmaceutical University, Nanjing 211198, China
| | - Tian-Jiao Zhou
- State Key Laboratory of Natural Medicines, China Pharmaceutical University, Nanjing 211198, China
| | - Yi Wang
- State Key Laboratory of Natural Medicines, China Pharmaceutical University, Nanjing 211198, China
| | - Ling-Feng Zhang
- State Key Laboratory of Natural Medicines, China Pharmaceutical University, Nanjing 211198, China
| | - Ling Li
- Department of Endocrinology, Zhongda Hospital, School of Medicine, Southeast University, Nanjing 210009, China.
| | - Chong-Su Cho
- Department of Agricultural Biotechnology and Research Institute for Agriculture and Life Sciences, Seoul National University, Seoul, 08826, Korea.
| | - Hu-Lin Jiang
- State Key Laboratory of Natural Medicines, China Pharmaceutical University, Nanjing 211198, China; College of Pharmacy, Yanbian University, Yanji 133002, China.
| |
Collapse
|
261
|
Shi M, Lu Y, Mohyeddin A, Qi F, Pan Y. Preservation of Eschar Prevents Excessive Wound Healing by Reducing M2 Macrophages Polarization. PLASTIC AND RECONSTRUCTIVE SURGERY-GLOBAL OPEN 2023; 11:e5238. [PMID: 37731728 PMCID: PMC10508428 DOI: 10.1097/gox.0000000000005238] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2023] [Accepted: 07/11/2023] [Indexed: 09/22/2023]
Abstract
Background Removal of the eschar has gradually become a consensus on treatments of deep dermal necrosis after skin trauma in recent years, whereas exaggerated scar contracture and tissue proliferation developed during healing have received little attention. Here, the authors investigated the effects of eschar on excessive wound healing of small dermal damage and focused on the role M2 macrophages played, hoping to offer a theoretical basis to improve patients' cosmetic satisfaction. Methods A mouse dorsal wound model (n = 12) was established by electric heating pads heating for 20 seconds on each side of the spine, and the left side was the preserved group. Macrophage numbers, expression of wound-healing-associated proteins, and inflammatory cytokine levels were assessed at different time points by immunohistochemistry and quantitative real-time polymerase chain reaction. A co-culture system of M2 macrophages and myofibroblasts was created in vitro. Immunohistochemistry, real-time polymerase chain reaction, and Western blot were performed to evaluate the proliferation, migration, and protein expression of myofibroblasts. Results Preserving eschar inhibited contraction-associated proteins (α-smooth muscle actin and vimentin) and collagen expression, inflammatory cytokine (IL-1β, IL-10, TFN-α, and IL-4) expression, and M2 macrophage infiltration. Mechanistically, M2 macrophages potentially contributed to excessive wound healing by promoting myofibroblasts proliferation, migration, and production of contraction-associated proteins. Conclusion Eschar preservation in wounds could reduce inflammation and negatively modulate myofibroblasts by inhibiting M2 macrophage polarization and infiltration, preventing excessive wound contraction and collagen deposition.
Collapse
Affiliation(s)
- Mingyue Shi
- From the Department of Plastic Surgery, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Yao Lu
- From the Department of Plastic Surgery, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Ali Mohyeddin
- From the Department of Plastic Surgery, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Fazhi Qi
- From the Department of Plastic Surgery, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Yuyan Pan
- From the Department of Plastic Surgery, Zhongshan Hospital, Fudan University, Shanghai, China
| |
Collapse
|
262
|
Yu L, Gao Y, Aaron N, Qiang L. A glimpse of the connection between PPARγ and macrophage. Front Pharmacol 2023; 14:1254317. [PMID: 37701041 PMCID: PMC10493289 DOI: 10.3389/fphar.2023.1254317] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2023] [Accepted: 07/31/2023] [Indexed: 09/14/2023] Open
Abstract
Nuclear receptors are ligand-regulated transcription factors that regulate vast cellular activities and serve as an important class of drug targets. Among them, peroxisome proliferator-activated receptors (PPARs) are members of the nuclear receptor family and have been extensively studied for their roles in metabolism, differentiation, development, and cancer, among others. Recently, there has been considerable interest in understanding and defining the function of PPARs and their agonists in regulating innate and adaptive immune responses and their pharmacological potential in combating chronic inflammatory diseases. In this review, we focus on emerging evidence for the potential role of PPARγ in macrophage biology, which is the prior innate immune executive in metabolic and tissue homeostasis. We also discuss the role of PPARγ as a regulator of macrophage function in inflammatory diseases. Lastly, we discuss the possible application of PPARγ antagonists in metabolic pathologies.
Collapse
Affiliation(s)
- Lexiang Yu
- Naomi Berrie Diabetes Center, Columbia University, New York, NY, United States
- Department of Pathology and Cell Biology, Columbia University, New York, NY, United States
- Department of Medicine, Vagelos College of Physicians and Surgeons, Columbia University, New York, NY, United States
| | - Yuen Gao
- Department of Physiology, Michigan State University, East Lansing, MI, United States
| | - Nicole Aaron
- Naomi Berrie Diabetes Center, Columbia University, New York, NY, United States
- Department of Molecular Pharmacology and Therapeutics, Columbia University, New York, NY, United States
| | - Li Qiang
- Naomi Berrie Diabetes Center, Columbia University, New York, NY, United States
- Department of Pathology and Cell Biology, Columbia University, New York, NY, United States
- Department of Medicine, Vagelos College of Physicians and Surgeons, Columbia University, New York, NY, United States
| |
Collapse
|
263
|
Hu Y, Xiong Y, Zhu Y, Zhou F, Liu X, Chen S, Li Z, Qi S, Chen L. Copper-Epigallocatechin Gallate Enhances Therapeutic Effects of 3D-Printed Dermal Scaffolds in Mitigating Diabetic Wound Scarring. ACS APPLIED MATERIALS & INTERFACES 2023; 15:38230-38246. [PMID: 37535406 PMCID: PMC10436249 DOI: 10.1021/acsami.3c04733] [Citation(s) in RCA: 18] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/03/2023] [Accepted: 07/10/2023] [Indexed: 08/04/2023]
Abstract
Morbid dermal templates, microangiopathy, and abnormal inflammation are the three most critical reasons for the scarred healing and the high recurrence rate of diabetic wounds. In this present study, a combination of a methacrylated decellularized extracellular matrix (ECMMA, aka EM)-based hydrogel system loaded with copper-epigallocatechin gallate (Cu-EGCG) capsules is proposed to fabricate bio-printed dermal scaffolds for diabetic wound treatment. Copper ions act as a bioactive element for promoting angiogenesis, and EGCG can inhibit inflammation on the wound site. In addition to the above activities, EM/Cu-EGCG (E/C) dermal scaffolds can also provide optimized templates and nutrient exchange space for guiding the orderly deposition and remodeling of ECM. In vitro experiments have shown that the E/C hydrogel can promote angiogenesis and inhibit the polarization of macrophages to the M1 pro-inflammatory phenotype. In the full-thickness skin defect model of diabetic rats, the E/C dermal scaffold combined with split-thickness skin graft transplantation can alleviate pathological scarring via promoting angiogenesis and driving macrophage polarization to the anti-inflammatory M2 phenotype. These may be attributed to the scaffold-actuated expression of angiogenesis-related genes in the HIF-1α/vascular endothelial growth factor pathway and decreased expression of inflammation-related genes in the TNF-α/NF-κB/MMP9 pathway. The results of this study show that the E/C dermal scaffold could serve as a promising artificial dermal analogue for solving the problems of delayed wound healing and reulceration of diabetic wounds.
Collapse
Affiliation(s)
- Yanke Hu
- Department
of Burn, Wound Repair & Reconstruction, The First Affiliated Hospital of Sun Yat-Sen University, Guangzhou, Guangdong 510080, China
- Guangdong
Provincial Engineering Technology Research Center of Burn and Wound
Accurate Diagnosis and Treatment Key Technology and Series of Products, Sun Yat-Sen University, Guangzhou, Guangdong 510080, China
- Institute
of Precision Medicine, The First Affiliated
Hospital, Sun Yat-Sen University, Guangzhou, Guangdong 510080, China
| | - Yahui Xiong
- Department
of Burn, Wound Repair & Reconstruction, The First Affiliated Hospital of Sun Yat-Sen University, Guangzhou, Guangdong 510080, China
- Guangdong
Provincial Engineering Technology Research Center of Burn and Wound
Accurate Diagnosis and Treatment Key Technology and Series of Products, Sun Yat-Sen University, Guangzhou, Guangdong 510080, China
- Institute
of Precision Medicine, The First Affiliated
Hospital, Sun Yat-Sen University, Guangzhou, Guangdong 510080, China
| | - Yongkang Zhu
- Department
of Burn, Wound Repair & Reconstruction, The First Affiliated Hospital of Sun Yat-Sen University, Guangzhou, Guangdong 510080, China
- Guangdong
Provincial Engineering Technology Research Center of Burn and Wound
Accurate Diagnosis and Treatment Key Technology and Series of Products, Sun Yat-Sen University, Guangzhou, Guangdong 510080, China
- Institute
of Precision Medicine, The First Affiliated
Hospital, Sun Yat-Sen University, Guangzhou, Guangdong 510080, China
| | - Fei Zhou
- Department
of Burn, Wound Repair & Reconstruction, The First Affiliated Hospital of Sun Yat-Sen University, Guangzhou, Guangdong 510080, China
- Guangdong
Provincial Engineering Technology Research Center of Burn and Wound
Accurate Diagnosis and Treatment Key Technology and Series of Products, Sun Yat-Sen University, Guangzhou, Guangdong 510080, China
- Institute
of Precision Medicine, The First Affiliated
Hospital, Sun Yat-Sen University, Guangzhou, Guangdong 510080, China
| | - Xiaogang Liu
- Department
of Burn, Wound Repair & Reconstruction, The First Affiliated Hospital of Sun Yat-Sen University, Guangzhou, Guangdong 510080, China
- Guangdong
Provincial Engineering Technology Research Center of Burn and Wound
Accurate Diagnosis and Treatment Key Technology and Series of Products, Sun Yat-Sen University, Guangzhou, Guangdong 510080, China
- Institute
of Precision Medicine, The First Affiliated
Hospital, Sun Yat-Sen University, Guangzhou, Guangdong 510080, China
| | - Shuying Chen
- Department
of Burn, Wound Repair & Reconstruction, The First Affiliated Hospital of Sun Yat-Sen University, Guangzhou, Guangdong 510080, China
- Guangdong
Provincial Engineering Technology Research Center of Burn and Wound
Accurate Diagnosis and Treatment Key Technology and Series of Products, Sun Yat-Sen University, Guangzhou, Guangdong 510080, China
- Institute
of Precision Medicine, The First Affiliated
Hospital, Sun Yat-Sen University, Guangzhou, Guangdong 510080, China
| | - Zhanpeng Li
- Department
of Burn, Wound Repair & Reconstruction, The First Affiliated Hospital of Sun Yat-Sen University, Guangzhou, Guangdong 510080, China
- Guangdong
Provincial Engineering Technology Research Center of Burn and Wound
Accurate Diagnosis and Treatment Key Technology and Series of Products, Sun Yat-Sen University, Guangzhou, Guangdong 510080, China
- Institute
of Precision Medicine, The First Affiliated
Hospital, Sun Yat-Sen University, Guangzhou, Guangdong 510080, China
| | - Shaohai Qi
- Department
of Burn, Wound Repair & Reconstruction, The First Affiliated Hospital of Sun Yat-Sen University, Guangzhou, Guangdong 510080, China
- Guangdong
Provincial Engineering Technology Research Center of Burn and Wound
Accurate Diagnosis and Treatment Key Technology and Series of Products, Sun Yat-Sen University, Guangzhou, Guangdong 510080, China
- Institute
of Precision Medicine, The First Affiliated
Hospital, Sun Yat-Sen University, Guangzhou, Guangdong 510080, China
| | - Lei Chen
- Department
of Burn, Wound Repair & Reconstruction, The First Affiliated Hospital of Sun Yat-Sen University, Guangzhou, Guangdong 510080, China
- Guangdong
Provincial Engineering Technology Research Center of Burn and Wound
Accurate Diagnosis and Treatment Key Technology and Series of Products, Sun Yat-Sen University, Guangzhou, Guangdong 510080, China
- Institute
of Precision Medicine, The First Affiliated
Hospital, Sun Yat-Sen University, Guangzhou, Guangdong 510080, China
| |
Collapse
|
264
|
Linnemann C, Şahin F, Li N, Pscherer S, Götz F, Histing T, Nussler AK, Ehnert S. Insulin Can Delay Neutrophil Extracellular Trap Formation In Vitro-Implication for Diabetic Wound Care? BIOLOGY 2023; 12:1082. [PMID: 37626968 PMCID: PMC10452400 DOI: 10.3390/biology12081082] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/29/2023] [Revised: 07/24/2023] [Accepted: 07/31/2023] [Indexed: 08/27/2023]
Abstract
Diabetes is a worldwide evolving disease with many associated complications, one of which is delayed or impaired wound healing. Appropriate wound healing strongly relies on the inflammatory reaction directly after injury, which is often altered in diabetic wound healing. After an injury, neutrophils are the first cells to enter the wound site. They have a special defense mechanism, neutrophil extracellular traps (NETs), consisting of released DNA coated with antimicrobial proteins and histones. Despite being a powerful weapon against pathogens, NETs were shown to contribute to impaired wound healing in diabetic mice and are associated with amputations in diabetic foot ulcer patients. The anti-diabetic drugs metformin and liraglutide have already been shown to regulate NET formation. In this study, the effect of insulin was investigated. NET formation after stimulation with PMA (phorbol myristate acetate), LPS (lipopolysaccharide), or calcium ionophore (CI) in the presence/absence of insulin was analyzed. Insulin led to a robust delay of LPS- and PMA-induced NET formation but had no effect on CI-induced NET formation. Mechanistically, insulin induced reactive oxygen species, phosphorylated p38, and ERK, but reduced citrullination of histone H3. Instead, bacterial killing was induced. Insulin might therefore be a new tool for the regulation of NET formation during diabetic wound healing, either in a systemic or topical application.
Collapse
Affiliation(s)
- Caren Linnemann
- Siegfried Weller Institute for Trauma Research, BG Unfallklinik Tübingen, Eberhard Karls Universität Tuebingen, 72076 Tuebingen, Germany; (C.L.); (A.K.N.)
| | - Filiz Şahin
- Siegfried Weller Institute for Trauma Research, BG Unfallklinik Tübingen, Eberhard Karls Universität Tuebingen, 72076 Tuebingen, Germany; (C.L.); (A.K.N.)
| | - Ningna Li
- Microbial Genetics, Interfaculty Institute of Microbiology and Infection Medicine, Eberhard Karls Universität Tuebingen, 72076 Tuebingen, Germany
| | - Stefan Pscherer
- Siegfried Weller Institute for Trauma Research, BG Unfallklinik Tübingen, Eberhard Karls Universität Tuebingen, 72076 Tuebingen, Germany; (C.L.); (A.K.N.)
- Department of Internal Medicine III, Sophien- and Hufeland-Hospital, 99425 Weimar, Germany
| | - Friedrich Götz
- Microbial Genetics, Interfaculty Institute of Microbiology and Infection Medicine, Eberhard Karls Universität Tuebingen, 72076 Tuebingen, Germany
| | - Tina Histing
- Siegfried Weller Institute for Trauma Research, BG Unfallklinik Tübingen, Eberhard Karls Universität Tuebingen, 72076 Tuebingen, Germany; (C.L.); (A.K.N.)
| | - Andreas K. Nussler
- Siegfried Weller Institute for Trauma Research, BG Unfallklinik Tübingen, Eberhard Karls Universität Tuebingen, 72076 Tuebingen, Germany; (C.L.); (A.K.N.)
| | - Sabrina Ehnert
- Siegfried Weller Institute for Trauma Research, BG Unfallklinik Tübingen, Eberhard Karls Universität Tuebingen, 72076 Tuebingen, Germany; (C.L.); (A.K.N.)
| |
Collapse
|
265
|
Illi B, Nasi S. Myc beyond Cancer: Regulation of Mammalian Tissue Regeneration. PATHOPHYSIOLOGY 2023; 30:346-365. [PMID: 37606389 PMCID: PMC10443299 DOI: 10.3390/pathophysiology30030027] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2023] [Revised: 07/28/2023] [Accepted: 07/31/2023] [Indexed: 08/23/2023] Open
Abstract
Myc is one of the most well-known oncogenes driving tumorigenesis in a wide variety of tissues. From the brain to blood, its deregulation derails physiological pathways that grant the correct functioning of the cell. Its action is carried out at the gene expression level, where Myc governs basically every aspect of transcription. Indeed, in addition to its role as a canonical, chromatin-bound transcription factor, Myc rules RNA polymerase II (RNAPII) transcriptional pause-release, elongation and termination and mRNA capping. For this reason, it is evident that minimal perturbations of Myc function mirror malignant cell behavior and, consistently, a large body of literature mainly focuses on Myc malfunctioning. In healthy cells, Myc controls molecular mechanisms involved in pivotal functions, such as cell cycle (and proliferation thereof), apoptosis, metabolism and cell size, angiogenesis, differentiation and stem cell self-renewal. In this latter regard, Myc has been found to also regulate tissue regeneration, a hot topic in the research fields of aging and regenerative medicine. Indeed, Myc appears to have a role in wound healing, in peripheral nerves and in liver, pancreas and even heart recovery. Herein, we discuss the state of the art of Myc's role in tissue regeneration, giving an overview of its potent action beyond cancer.
Collapse
Affiliation(s)
- Barbara Illi
- Institute of Molecular Biology and Pathology, National Research Council, c/o Department of Biology and Biotechnology “Charles Darwin”, Sapienza University of Rome, 00185 Rome, Italy
| | - Sergio Nasi
- Institute of Molecular Biology and Pathology, National Research Council, c/o Department of Biology and Biotechnology “Charles Darwin”, Sapienza University of Rome, 00185 Rome, Italy
| |
Collapse
|
266
|
Li Y, Zhong Z, Xu C, Wu X, Li J, Tao W, Wang J, Du Y, Zhang S. 3D micropattern force triggers YAP nuclear entry by transport across nuclear pores and modulates stem cells paracrine. Natl Sci Rev 2023; 10:nwad165. [PMID: 37457331 PMCID: PMC10347367 DOI: 10.1093/nsr/nwad165] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2023] [Revised: 04/27/2023] [Accepted: 05/25/2023] [Indexed: 07/18/2023] Open
Abstract
Biophysical cues of the cellular microenvironment tremendously influence cell behavior by mechanotransduction. However, it is still unclear how cells sense and transduce the mechanical signals from 3D geometry to regulate cell function. Here, the mechanotransduction of human mesenchymal stem cells (MSCs) triggered by 3D micropatterns and its effect on the paracrine of MSCs are systematically investigated. Our findings show that 3D micropattern force could influence the spatial reorganization of the cytoskeleton, leading to different local forces which mediate nucleus alteration such as orientation, morphology, expression of Lamin A/C and chromatin condensation. Specifically, in the triangular prism and cuboid micropatterns, the ordered F-actin fibers are distributed over and fully transmit compressive forces to the nucleus, which results in nuclear flattening and stretching of nuclear pores, thus enhancing the nuclear import of YES-associated protein (YAP). Furthermore, the activation of YAP significantly enhances the paracrine of MSCs and upregulates the secretion of angiogenic growth factors. In contrast, the fewer compressive forces on the nucleus in cylinder and cube micropatterns cause less YAP entering the nucleus. The skin repair experiment provides the first in vivo evidence that enhanced MSCs paracrine by 3D geometry significantly promotes tissue regeneration. The current study contributes to understanding the in-depth mechanisms of mechanical signals affecting cell function and provides inspiration for innovative design of biomaterials.
Collapse
Affiliation(s)
| | | | - Cunjing Xu
- Advanced Biomaterials and Tissue Engineering Center, Huazhong University of Science and Technology, Wuhan430074, China
- Department of Biomedical Engineering, Huazhong University of Science and Technology, Wuhan430074, China
| | - Xiaodan Wu
- Advanced Biomaterials and Tissue Engineering Center, Huazhong University of Science and Technology, Wuhan430074, China
- Department of Biomedical Engineering, Huazhong University of Science and Technology, Wuhan430074, China
| | - Jiaqi Li
- Advanced Biomaterials and Tissue Engineering Center, Huazhong University of Science and Technology, Wuhan430074, China
- Department of Biomedical Engineering, Huazhong University of Science and Technology, Wuhan430074, China
| | - Weiyong Tao
- Advanced Biomaterials and Tissue Engineering Center, Huazhong University of Science and Technology, Wuhan430074, China
- Department of Biomedical Engineering, Huazhong University of Science and Technology, Wuhan430074, China
| | - Jianglin Wang
- Advanced Biomaterials and Tissue Engineering Center, Huazhong University of Science and Technology, Wuhan430074, China
- Department of Biomedical Engineering, Huazhong University of Science and Technology, Wuhan430074, China
| | | | | |
Collapse
|
267
|
Lin Z, Chen Z, Chen Y, Yang N, Shi J, Tang Z, Zhang C, Lin H, Yin J. Hydrogenated silicene nanosheet functionalized scaffold enables immuno-bone remodeling. EXPLORATION (BEIJING, CHINA) 2023; 3:20220149. [PMID: 37933236 PMCID: PMC10624372 DOI: 10.1002/exp.20220149] [Citation(s) in RCA: 20] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 11/01/2022] [Accepted: 04/03/2023] [Indexed: 11/08/2023]
Abstract
An ideal implant needs to have the ability to coordinate the foreign body response and tissue regeneration. Here, Hydrogenated-silicon nanosheets (H-Si NSs) with favorable biodegradability are integrated and functionalized into a β-tricalcium phosphate scaffold (H-Si TCP) for bone defect healing. H-Si TCP can greatly improve bone regeneration through osteoimmunomodulation-guided biodegradation in vivo. The spatiotemporal regulation of degradation products replenishes sufficient nutrients step by step for the entire process of bone repair. Extracellular and intracellular reactive oxygen species (ROS) are first downregulated by reaction with H-Si NSs, followed by marked M2 polarization, remodeling the micro-environment timely for immune-bone regeneration. The release of primary reaction products awakened bone marrow mesenchymal stem cells (BMSCs), which are converted into osteoblasts anchored on scaffolds. Subsequently, biomineralization is promoted by the final degradation products. The intrinsic ROS-responsive, immunoregulatory, and osteo-promotive capability of 2D H-Si NSs makes such composite H-Si TCP scaffold a highly potential alternative for the treatment of critical bone defect.
Collapse
Affiliation(s)
- Zixuan Lin
- Institute of Microsurgery on ExtremitiesDepartment of Orthopaedic SurgeryShanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of MedicineShanghaiP. R. China
| | - Zhixin Chen
- State Key Laboratory of High Performance Ceramics and Superfine MicrostructureShanghai Institute of Ceramics Chinese Academy of SciencesShanghaiP. R. China
| | - Yiwei Chen
- Institute of Microsurgery on ExtremitiesDepartment of Orthopaedic SurgeryShanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of MedicineShanghaiP. R. China
| | - Nan Yang
- Institute of Microsurgery on ExtremitiesDepartment of Orthopaedic SurgeryShanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of MedicineShanghaiP. R. China
| | - Jianlin Shi
- State Key Laboratory of High Performance Ceramics and Superfine MicrostructureShanghai Institute of Ceramics Chinese Academy of SciencesShanghaiP. R. China
- Shanghai Tenth People's HospitalShanghai Frontiers Science Center of Nanocatalytic MedicineSchool of MedicineTongji UniversityShanghaiP. R. China
| | - Zhongmin Tang
- Departments of Radiology and Medical PhysicsUniversity of Wisconsin‐MadisonMadisonWisconsinUSA
| | - Changqing Zhang
- Institute of Microsurgery on ExtremitiesDepartment of Orthopaedic SurgeryShanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of MedicineShanghaiP. R. China
| | - Han Lin
- State Key Laboratory of High Performance Ceramics and Superfine MicrostructureShanghai Institute of Ceramics Chinese Academy of SciencesShanghaiP. R. China
- Shanghai Tenth People's HospitalShanghai Frontiers Science Center of Nanocatalytic MedicineSchool of MedicineTongji UniversityShanghaiP. R. China
| | - Junhui Yin
- Institute of Microsurgery on ExtremitiesDepartment of Orthopaedic SurgeryShanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of MedicineShanghaiP. R. China
| |
Collapse
|
268
|
Torres Costa KC, Santana Vieira Santos V, Rezende Vaz E, Natalie Cirilo Gimenes S, Ian Veloso Correia L, Brito de Souza J, de Almeida Araújo Santos F, de Melo Rodrigues V, Ricardo Goulart L, Alonso Goulart V. A novel peptide able to reduce PLA 2 activity and modulate inflammatory cytokine production. Toxicon 2023; 231:107207. [PMID: 37364619 DOI: 10.1016/j.toxicon.2023.107207] [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: 04/11/2023] [Revised: 05/19/2023] [Accepted: 06/22/2023] [Indexed: 06/28/2023]
Abstract
Phospholipases A2 (PLA2s) are associated with inflammatory response, performing a complex process involving, specially, cytokines. The excess of pro-inflammatory cytokines induces a chronic inflammatory response and can cause several disorders in the body. Therefore, the inhibition or regulation of cytokines' signaling pathways is a target for new treatment development strategies. Thus, this study aimed to select PLA2 inhibitor mimetic peptides through phage display technology with anti-inflammatory activity. Specific mimetic peptides were selected using BpPLA2-TXI, a PLA2 isolated from Bothrops pauloensis, as a target, and γCdcPL, a PLA2 inhibitor isolated from Crotalus durissus collilineatus, which was used as a competitor during the elution step. We selected the peptide C2PD, which seems to play a pivotal role in the modulation of IL-6, IL-1β, and IL-10 cytokines in inflammatory cells. The C2PD showed a significant reduction in PLA2 activity. Furthermore, the synthetic peptide was tested in PBMC and showed a significant down-modulation of IL-6 and IL-1β release, whereas IL-10 responses were up-regulated. Our findings suggest that this novel peptide may be a potential therapeutic candidate for the treatment of inflammatory diseases, mainly due to its anti-inflammatory properties and absence of cytotoxicity.
Collapse
Affiliation(s)
- Kellen Cristina Torres Costa
- Laboratory of Nanobiotechnology Prof. Dr. Luiz Ricardo Goulart Filho, Institute of Biotechnology, Federal University of Uberlândia, Umuarama Campus, Avenida Pará, 1720, 38.400-902, Minas Gerais, Brazil.
| | - Vanessa Santana Vieira Santos
- Laboratory of Environmental Health, Department of Environmental Health, Institute of Biotechnology, Federal University of Uberlandia, Santa Monica Campus, Avenida João Naves de Ávila, 2121, 38.408-100, Uberlandia, Minas Gerais, Brazil
| | - Emília Rezende Vaz
- Laboratory of Nanobiotechnology Prof. Dr. Luiz Ricardo Goulart Filho, Institute of Biotechnology, Federal University of Uberlândia, Umuarama Campus, Avenida Pará, 1720, 38.400-902, Minas Gerais, Brazil
| | | | - Lucas Ian Veloso Correia
- Laboratory of Nanobiotechnology Prof. Dr. Luiz Ricardo Goulart Filho, Institute of Biotechnology, Federal University of Uberlândia, Umuarama Campus, Avenida Pará, 1720, 38.400-902, Minas Gerais, Brazil
| | - Jessica Brito de Souza
- Laboratory of Nanobiotechnology Prof. Dr. Luiz Ricardo Goulart Filho, Institute of Biotechnology, Federal University of Uberlândia, Umuarama Campus, Avenida Pará, 1720, 38.400-902, Minas Gerais, Brazil
| | - Fabiana de Almeida Araújo Santos
- Laboratory of Nanobiotechnology Prof. Dr. Luiz Ricardo Goulart Filho, Institute of Biotechnology, Federal University of Uberlândia, Umuarama Campus, Avenida Pará, 1720, 38.400-902, Minas Gerais, Brazil
| | - Veridiana de Melo Rodrigues
- Laboratory of Nanobiotechnology Prof. Dr. Luiz Ricardo Goulart Filho, Institute of Biotechnology, Federal University of Uberlândia, Umuarama Campus, Avenida Pará, 1720, 38.400-902, Minas Gerais, Brazil; Laboratory of Biochemistry and Animal Toxins, Institute of Biotechnology, Federal University of Uberlândia, Umuarama Campus, Avenida Pará, 1720, 38.400-902, Minas Gerais, Brazil
| | - Luiz Ricardo Goulart
- Laboratory of Nanobiotechnology Prof. Dr. Luiz Ricardo Goulart Filho, Institute of Biotechnology, Federal University of Uberlândia, Umuarama Campus, Avenida Pará, 1720, 38.400-902, Minas Gerais, Brazil
| | - Vivian Alonso Goulart
- Laboratory of Nanobiotechnology Prof. Dr. Luiz Ricardo Goulart Filho, Institute of Biotechnology, Federal University of Uberlândia, Umuarama Campus, Avenida Pará, 1720, 38.400-902, Minas Gerais, Brazil
| |
Collapse
|
269
|
Bogen KT. Ultrasensitive dose-response for asbestos cancer risk implied by new inflammation-mutation model. ENVIRONMENTAL RESEARCH 2023; 230:115047. [PMID: 36965808 DOI: 10.1016/j.envres.2022.115047] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/19/2022] [Accepted: 12/09/2022] [Indexed: 05/30/2023]
Abstract
Alterations in complex cellular phenotype each typically involve multistep activation of an ultrasensitive molecular switch (e.g., to adaptively initiate an apoptosis, inflammasome, Nrf2-ARE anti-oxidant, or heat-shock activation pathway) that triggers expression of a suite of target genes while efficiently limiting false-positive switching from a baseline state. Such switches exhibit nonlinear signal-activation relationships. In contrast, a linear no-threshold (LNT) dose-response relationship is expected for damage that accumulates in proportion to dose, as hypothesized for increased risk of cancer in relation to genotoxic dose according to the multistage somatic mutation/clonal-expansion theory of cancer, e.g., as represented in the Moolgavkar-Venzon-Knudsen (MVK) cancer model by a doubly stochastic nonhomogeneous Poisson process. Mesothelioma and lung cancer induced by exposure to carcinogenic (e.g., certain asbestos) fibers in humans and experimental animals are thought to involve modes of action driven by mutations, cytotoxicity-associated inflammation, or both, rendering ambiguous expectations concerning the nature of model-implied shape of the low-dose response for above-background increase in risk of incurring these endpoints. A recent Inflammation Somatic Mutation (ISM) theory of cancer posits instead that tissue-damage-associated inflammation that epigenetically recruits, activates and orchestrates stem cells to engage in tissue repair does not merely promote cancer, but rather is a requisite co-initiator (acting together with as few as two somatic mutations) of the most efficient pathway to any type of cancer in any reparable tissue (Dose-Response 2019; 17(2):1-12). This theory is reviewed, implications of this theory are discussed in relation to mesothelioma and lung cancer associated with chronic asbestos inhalation, one of the two types of ISM-required mutations is here hypothesized to block or impede inflammation resolution (e.g., by doing so for GPCR-mediated signal transduction by one or more endogenous autacoid specialized pro-resolving mediators or SPMs), and supporting evidence for this hypothesis is discussed.
Collapse
Affiliation(s)
- Kenneth T Bogen
- 9832 Darcy Forest Drive, Silver Spring, MD, 20910, United States.
| |
Collapse
|
270
|
Wei H, Yu S, Zhang Y, Zhang H, Ma Y, Xu M, An P, Zhou Y, Halila S, Wei Y, Chen J. Injectable chitosan/xyloglucan composite hydrogel with mechanical adaptivity and endogenous bioactivity for skin repair. Carbohydr Polym 2023; 313:120904. [PMID: 37182937 DOI: 10.1016/j.carbpol.2023.120904] [Citation(s) in RCA: 18] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2022] [Revised: 03/17/2023] [Accepted: 04/09/2023] [Indexed: 05/16/2023]
Abstract
Delayed or chronic wound healing is one of severe clinical issues. Developing scaffold materials capable of supporting cells and inducing tissue regeneration remains a challenge. Here, a polysaccharide-based hydrogel is constructed for promoting full-thickness skin wound healing in mouse model. The engineering hydrogel consists of a dynamic crosslinking network formed by the Schiff base reaction between aldehyde-containing xyloglucan and methacrylated chitosan. Its reversible gel-sol-gel transition upon shearing force is highly beneficial to completely cover and fill irregular wound shape. The second covalent cross-linking network achieved by photo-initiated polymerization offers a feasible way to tune the mechanical property of hydrogel after injection, with an ideal mechanical adaptivity for clinical application. Remarkably, both in vitro and in vivo evaluations demonstrate that the hydrogel with endogenously bioactive galactoside units can promote cell spheroid formation and accelerate wound healing by expediting re-epithelialization, collagen deposition, angiogenesis as well as the formation of hair follicles.
Collapse
Affiliation(s)
- Hua Wei
- Key Laboratory of Colloid and Interface Chemistry, Ministry of Education, School of Chemistry and Chemical Engineering, Shandong University, Jinan 250100, China
| | - Shengkai Yu
- Research Institute for Medical and Biological Engineering, Ningbo University, Ningbo 315211, China
| | - Yansheng Zhang
- Zhejiang Engineering Research Center for Biomedical Materials, Zhejiang International Scientific and Technological Cooperative Base of Biomedical Materials and Technology, Cixi Institute of Biomedical Engineering, Ningbo Institute of Materials Technology and Engineering, Chinese Academy of Sciences, Ningbo 315300, China; University of Chinese Academy of Sciences, Beijing 100049, China
| | - Hua Zhang
- Research Institute for Medical and Biological Engineering, Ningbo University, Ningbo 315211, China
| | - Yuxi Ma
- Zhejiang Engineering Research Center for Biomedical Materials, Zhejiang International Scientific and Technological Cooperative Base of Biomedical Materials and Technology, Cixi Institute of Biomedical Engineering, Ningbo Institute of Materials Technology and Engineering, Chinese Academy of Sciences, Ningbo 315300, China; University of Chinese Academy of Sciences, Beijing 100049, China
| | - Mengjia Xu
- Zhejiang Engineering Research Center for Biomedical Materials, Zhejiang International Scientific and Technological Cooperative Base of Biomedical Materials and Technology, Cixi Institute of Biomedical Engineering, Ningbo Institute of Materials Technology and Engineering, Chinese Academy of Sciences, Ningbo 315300, China
| | - Peng An
- Zhejiang Engineering Research Center for Biomedical Materials, Zhejiang International Scientific and Technological Cooperative Base of Biomedical Materials and Technology, Cixi Institute of Biomedical Engineering, Ningbo Institute of Materials Technology and Engineering, Chinese Academy of Sciences, Ningbo 315300, China
| | - Yang Zhou
- Zhejiang Engineering Research Center for Biomedical Materials, Zhejiang International Scientific and Technological Cooperative Base of Biomedical Materials and Technology, Cixi Institute of Biomedical Engineering, Ningbo Institute of Materials Technology and Engineering, Chinese Academy of Sciences, Ningbo 315300, China
| | - Sami Halila
- Université Grenoble Alpes, CNRS, CERMAV, Grenoble 38000, France.
| | - Yen Wei
- Department of Chemistry and the Tsinghua Center for Frontier Polymer Research, Tsinghua University, Beijing 100084, China.
| | - Jing Chen
- Institute of Medical Sciences, The Second Hospital and Shandong University Center for Orthopaedics, Cheeloo College of Medicine, Shandong University, Jinan 250033, China.
| |
Collapse
|
271
|
Xu Y, Ying L, Lang JK, Hinz B, Zhao R. Modeling Mechanical Activation of Macrophages During Pulmonary Fibrogenesis for Targeted Anti-Fibrosis Therapy. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.07.19.549794. [PMID: 37503121 PMCID: PMC10370161 DOI: 10.1101/2023.07.19.549794] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 07/29/2023]
Abstract
Pulmonary fibrosis, as seen in idiopathic pulmonary fibrosis (IPF) and COVID-induced pulmonary fibrosis, is an often-fatal lung disease. Increased numbers of immune cells such as macrophages were shown to accumulate in the fibrotic lung, but it is unclear how they contribute to the development of fibrosis. To recapitulate the macrophage mechanical activation in the fibrotic lung tissue microenvironment, we developed a fibrotic microtissue model with cocultured human macrophages and fibroblasts. We show that profibrotic macrophages seeded on topographically controlled stromal tissue constructs become mechanically activated. The resulting co-alignment of macrophages, collagen fibers and fibroblasts promote widespread fibrogenesis in micro-engineered lung tissues. Anti-fibrosis treatment using pirfenidone disrupts the polarization and mechanical activation of profibrotic macrophages, leading to fibrosis inhibition. Pirfenidone inhibits the mechanical activation of macrophages by suppressing integrin αMβ2 (CD11b/CD18) and Rho-associated kinase 2, which is a previously unknown mechanism of action of the drug. Together, these results demonstrate a potential pulmonary fibrogenesis mechanism at the tissue level contributed by mechanically activated macrophages. We propose the coculture, force-sensing microtissue model as a powerful tool to study the complex immune-stromal cell interactions and the mechanism of action of anti-fibrosis drugs.
Collapse
|
272
|
Chu X, Xiong Y, Knoedler S, Lu L, Panayi AC, Alfertshofer M, Jiang D, Rinkevich Y, Lin Z, Zhao Z, Dai G, Mi B, Liu G. Immunomodulatory Nanosystems: Advanced Delivery Tools for Treating Chronic Wounds. RESEARCH (WASHINGTON, D.C.) 2023; 6:0198. [PMID: 37456931 PMCID: PMC10348408 DOI: 10.34133/research.0198] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 05/26/2023] [Accepted: 07/02/2023] [Indexed: 07/18/2023]
Abstract
The increasingly aging society led to a rise in the prevalence of chronic wounds (CWs), posing a significant burden to public health on a global scale. One of the key features of CWs is the presence of a maladjusted immune microenvironment characterized by persistent and excessive (hyper)inflammation. A variety of immunomodulatory therapies have been proposed to address this condition. Yet, to date, current delivery systems for immunomodulatory therapy remain inadequate and lack efficiency. This highlights the need for new therapeutic delivery systems, such as nanosystems, to manage the pathological inflammatory imbalance and, ultimately, improve the treatment outcomes of CWs. While a plethora of immunomodulatory nanosystems modifying the immune microenvironment of CWs have shown promising therapeutic effects, the literature on the intersection of immunomodulatory nanosystems and CWs remains relatively scarce. Therefore, this review aims to provide a comprehensive overview of the pathogenesis and characteristics of the immune microenvironment in CWs, discuss important advancements in our understanding of CW healing, and delineate the versatility and applicability of immunomodulatory nanosystems-based therapies in the therapeutic management of CWs. In addition, we herein also shed light on the main challenges and future perspectives in this rapidly evolving research field.
Collapse
Affiliation(s)
- Xiangyu Chu
- Department of Orthopedics, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China
- Hubei Province Key Laboratory of Oral and Maxillofacial Development and Regeneration, Wuhan 430022, China
| | - Yuan Xiong
- Department of Orthopedics, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China
- Hubei Province Key Laboratory of Oral and Maxillofacial Development and Regeneration, Wuhan 430022, China
| | - Samuel Knoedler
- Division of Plastic Surgery, Brigham and Women's Hospital, Harvard Medical School, Boston, MA 02152, USA
- Institute of Regenerative Biology and Medicine, Helmholtz Zentrum München, Max-Lebsche-Platz 31, 81377 Munich, Germany
| | - Li Lu
- Department of Orthopedics, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China
- Hubei Province Key Laboratory of Oral and Maxillofacial Development and Regeneration, Wuhan 430022, China
| | - Adriana C Panayi
- Division of Plastic Surgery, Brigham and Women's Hospital, Harvard Medical School, Boston, MA 02152, USA
- Department of Hand, Plastic and Reconstructive Surgery, Microsurgery, Burn Center, BG Trauma Center Ludwigshafen, University of Heidelberg, Ludwig-Guttmann-Strasse 13, 67071 Ludwigshafen/Rhine, Germany
| | - Michael Alfertshofer
- Division of Hand, Plastic and Aesthetic Surgery, Ludwig - Maximilian University Munich, Munich, Germany
| | - Dongsheng Jiang
- Institute of Regenerative Biology and Medicine, Helmholtz Zentrum München, Max-Lebsche-Platz 31, 81377 Munich, Germany
| | - Yuval Rinkevich
- Institute of Regenerative Biology and Medicine, Helmholtz Zentrum München, Max-Lebsche-Platz 31, 81377 Munich, Germany
| | - Ze Lin
- Department of Orthopedics, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China
- Hubei Province Key Laboratory of Oral and Maxillofacial Development and Regeneration, Wuhan 430022, China
| | - Zhiming Zhao
- Department of Orthopedics, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China
- Department of Orthopedics, Suizhou Hospital, Hubei University of Medicine, Suizhou 441300, China
| | - Guandong Dai
- Pingshan District People's Hospital of Shenzhen, Pingshan General Hospital of Southern Medical University, Shenzhen, Guangdong 518118, China
| | - Bobin Mi
- Department of Orthopedics, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China
- Hubei Province Key Laboratory of Oral and Maxillofacial Development and Regeneration, Wuhan 430022, China
| | - Guohui Liu
- Department of Orthopedics, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China
- Hubei Province Key Laboratory of Oral and Maxillofacial Development and Regeneration, Wuhan 430022, China
| |
Collapse
|
273
|
Qiu B, Lawan A, Xirouchaki CE, Yi JS, Robert M, Zhang L, Brown W, Fernández-Hernando C, Yang X, Tiganis T, Bennett AM. MKP1 promotes nonalcoholic steatohepatitis by suppressing AMPK activity through LKB1 nuclear retention. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.07.10.548263. [PMID: 37502892 PMCID: PMC10369865 DOI: 10.1101/2023.07.10.548263] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 07/29/2023]
Abstract
Nonalcoholic steatohepatitis (NASH) is triggered by hepatocyte death through activation of caspase 6, as a result of decreased adenosine monophosphate (AMP)-activated protein kinase-alpha (AMPKα) activity. Increased hepatocellular death promotes inflammation which drives hepatic fibrosis. We show that the nuclear-localized mitogen-activated protein kinase (MAPK) phosphatase-1 (MKP1) is upregulated in NASH patients and in NASH diet fed mice. The focus of this work was to investigate whether and how MKP1 is involved in the development of NASH. Under NASH conditions increased oxidative stress, induces MKP1 expression leading to nuclear p38 MAPK dephosphorylation and decreased liver kinase B1 (LKB1) phosphorylation at a site required to promote LKB1 nuclear exit. Hepatic deletion of MKP1 in NASH diet fed mice released nuclear LKB1 into the cytoplasm to activate AMPKα and prevent hepatocellular death, inflammation and NASH. Hence, nuclear-localized MKP1-p38 MAPK-LKB1 signaling is required to suppress AMPKα which triggers hepatocyte death and the development of NASH.
Collapse
|
274
|
Nalesso PRL, Vedovatto M, Gregório JES, Huang B, Vyas C, Santamaria-Jr M, Bártolo P, Caetano GF. Early In Vivo Osteogenic and Inflammatory Response of 3D Printed Polycaprolactone/Carbon Nanotube/Hydroxyapatite/Tricalcium Phosphate Composite Scaffolds. Polymers (Basel) 2023; 15:2952. [PMID: 37447597 DOI: 10.3390/polym15132952] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2023] [Revised: 06/29/2023] [Accepted: 07/03/2023] [Indexed: 07/15/2023] Open
Abstract
The development of advanced biomaterials and manufacturing processes to fabricate biologically and mechanically appropriate scaffolds for bone tissue is a significant challenge. Polycaprolactone (PCL) is a biocompatible and degradable polymer used in bone tissue engineering, but it lacks biofunctionalization. Bioceramics, such as hydroxyapatite (HA) and β tricalcium phosphate (β-TCP), which are similar chemically to native bone, can facilitate both osteointegration and osteoinduction whilst improving the biomechanics of a scaffold. Carbon nanotubes (CNTs) display exceptional electrical conductivity and mechanical properties. A major limitation is the understanding of how PCL-based scaffolds containing HA, TCP, and CNTs behave in vivo in a bone regeneration model. The objective of this study was to evaluate the use of three-dimensional (3D) printed PCL-based composite scaffolds containing CNTs, HA, and β-TCP during the initial osteogenic and inflammatory response phase in a critical bone defect rat model. Gene expression related to early osteogenesis, the inflammatory phase, and tissue formation was evaluated using quantitative real-time PCR (RT-qPCR). Tissue formation and mineralization were assessed by histomorphometry. The CNT+HA/TCP group presented higher expression of osteogenic genes after seven days. The CNT+HA and CNT+TCP groups stimulated higher gene expression for tissue formation and mineralization, and pro- and anti-inflammatory genes after 14 and 30 days. Moreover, the CNT+TCP and CNT+HA/TCP groups showed higher gene expressions related to M1 macrophages. The association of CNTs with ceramics at 10wt% (CNT+HA/TCP) showed lower expressions of inflammatory genes and higher osteogenic, presenting a positive impact and balanced cell signaling for early bone formation. The association of CNTs with both ceramics promoted a minor inflammatory response and faster bone tissue formation.
Collapse
Affiliation(s)
- Paulo Roberto Lopes Nalesso
- Graduate Program in Biomedical Sciences, University Centre of Hermínio Ometto Foundation, Araras 13607-339, SP, Brazil
| | - Matheus Vedovatto
- Graduate Program in Biomedical Sciences, University Centre of Hermínio Ometto Foundation, Araras 13607-339, SP, Brazil
| | | | - Boyang Huang
- Singapore Centre for 3D Printing, School of Mechanical and Aerospace Engineering, Nanyang Technological University, Jurong West, Singapore 639798, Singapore
| | - Cian Vyas
- Singapore Centre for 3D Printing, School of Mechanical and Aerospace Engineering, Nanyang Technological University, Jurong West, Singapore 639798, Singapore
- School of Mechanical, Aerospace and Civil Engineering, The University of Manchester, Manchester M13 9PL, UK
| | - Milton Santamaria-Jr
- Graduate Program of Orthodontics, University Centre of Hermínio Ometto Foundation, Araras 13607-339, SP, Brazil
- Department of Social and Pediatric Dentistry, UNESP - São Paulo State University, Institute of Science and Technology - College of Dentistry, São José dos Campos 12245-000, SP, Brazil
| | - Paulo Bártolo
- Singapore Centre for 3D Printing, School of Mechanical and Aerospace Engineering, Nanyang Technological University, Jurong West, Singapore 639798, Singapore
- School of Mechanical, Aerospace and Civil Engineering, The University of Manchester, Manchester M13 9PL, UK
| | - Guilherme Ferreira Caetano
- Graduate Program in Biomedical Sciences, University Centre of Hermínio Ometto Foundation, Araras 13607-339, SP, Brazil
- Graduate Program of Orthodontics, University Centre of Hermínio Ometto Foundation, Araras 13607-339, SP, Brazil
- Division of Dermatology, Department of Internal Medicine, Ribeirão Preto Medical School, São Paulo University (USP), Ribeirão Preto 14049-900, SP, Brazil
| |
Collapse
|
275
|
Perveen I, Bukhari B, Najeeb M, Nazir S, Faridi TA, Farooq M, Ahmad QUA, Abusalah MAHA, ALjaraedah TY, Alraei WY, Rabaan AA, Singh KKB, Abusalah MAHA. Hydrogen Therapy and Its Future Prospects for Ameliorating COVID-19: Clinical Applications, Efficacy, and Modality. Biomedicines 2023; 11:1892. [PMID: 37509530 PMCID: PMC10377251 DOI: 10.3390/biomedicines11071892] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2023] [Revised: 06/09/2023] [Accepted: 06/13/2023] [Indexed: 07/30/2023] Open
Abstract
Molecular hydrogen is renowned as an odorless and colorless gas. The recommendations developed by China suggest that the inhalation of hydrogen molecules is currently advised in COVID-19 pneumonia treatment. The therapeutic effects of molecular hydrogens have been confirmed after numerous clinical trials and animal-model-based experiments, which have expounded that the low molecular weight of hydrogen enables it to easily diffuse and permeate through the cell membranes to produce a variety of biological impacts. A wide range of both chronic and acute inflammatory diseases, which may include sepsis, pancreatitis, respiratory disorders, autoimmune diseases, ischemia-reperfusion damages, etc. may be treated and prevented by using it. H2 can primarily be inoculated through inhalation, by drinking water (which already contains H2), or by administrating the injection of saline H2 in the body. It may play a pivotal role as an antioxidant, in regulating the immune system, in anti-inflammatory activities (mitochondrial energy metabolism), and cell death (apoptosis, pyroptosis, and autophagy) by reducing the formation of excessive reactive O2 species and modifying the transcription factors in the nuclei of the cells. However, the fundamental process of molecular hydrogen is still not entirely understood. Molecular hydrogen H2 has a promising future in therapeutics based on its safety and possible usefulness. The current review emphasizes the antioxidative, anti-apoptotic, and anti-inflammatory effects of hydrogen molecules along with the underlying principle and fundamental mechanism involved, with a prime focus on the coronavirus disease of 2019 (COVID-19). This review will also provide strategies and recommendations for the therapeutic and medicinal applications of the hydrogen molecule.
Collapse
Affiliation(s)
- Ishrat Perveen
- Food and Biotechnology Research Centre, Pakistan Council of Scientific and Industrial Research Centre, Lahore 54590, Pakistan
| | - Bakhtawar Bukhari
- Food and Biotechnology Research Centre, Pakistan Council of Scientific and Industrial Research Centre, Lahore 54590, Pakistan
| | - Mahwish Najeeb
- University Institute of Public Health, The University of Lahore, Lahore 54590, Pakistan
| | - Sumbal Nazir
- School of Zoology, Minhaj University Lahore, Lahore 54770, Pakistan
| | - Tallat Anwar Faridi
- University Institute of Public Health, The University of Lahore, Lahore 54590, Pakistan
| | - Muhammad Farooq
- Food and Biotechnology Research Centre, Pakistan Council of Scientific and Industrial Research Centre, Lahore 54590, Pakistan
| | - Qurat-Ul-Ain Ahmad
- Division of Science and Technology, University of Education, Township Lahore, Lahore 54770, Pakistan
| | - Manal Abdel Haleem A Abusalah
- Department of Medical Microbiology and Parasitology, School of Medical Sciences, Universiti Sains Malaysia, Kubang Kerian 16150, Malaysia
| | - Thana' Y ALjaraedah
- Department of Diet Therapy Technology & Dietetics, Faculty of Allied Medical Sciences, Zarqa University, Al-Zarqa 13132, Jordan
| | - Wesal Yousef Alraei
- Department of Diet Therapy Technology & Dietetics, Faculty of Allied Medical Sciences, Zarqa University, Al-Zarqa 13132, Jordan
| | - Ali A Rabaan
- Molecular Diagnostic Laboratory, Johns Hopkins Aramco Healthcare, Dhahran 31311, Saudi Arabia
| | - Kirnpal Kaur Banga Singh
- Department of Medical Microbiology and Parasitology, School of Medical Sciences, Universiti Sains Malaysia, Kubang Kerian 16150, Malaysia
| | - Mai Abdel Haleem A Abusalah
- Department of Medical Laboratory Sciences, Faculty of Allied Medical Sciences, Zarqa University, Al-Zarqa 13132, Jordan
| |
Collapse
|
276
|
Di Fazio P, Mielke S, Böhm IT, Buchholz M, Matrood S, Schuppan D, Wissniowski T. Toll-like receptor 5 tunes hepatic and pancreatic stellate cells activation. BMJ Open Gastroenterol 2023; 10:e001148. [PMID: 37433685 PMCID: PMC10347502 DOI: 10.1136/bmjgast-2023-001148] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/22/2023] [Accepted: 06/21/2023] [Indexed: 07/13/2023] Open
Abstract
OBJECTIVE Stellate cells are responsible for liver and pancreas fibrosis and strictly correlate with tumourigenesis. Although their activation is reversible, an exacerbated signalling triggers chronic fibrosis. Toll-like receptors (TLRs) modulate stellate cells transition. TLR5 transduces the signal deriving by the binding to bacterial flagellin from invading mobile bacteria. DESIGN Human hepatic and pancreatic stellate cells were activated by the administration of transforming growth factor-beta (TGF-β). TLR5 was transiently knocked down by short-interference RNA transfection. Reverse Transcription-quantitativePCR and western blot were performed to analyse the transcript and protein level of TLR5 and the transition players. Fluorescence microscopy was performed to identify these targets in spheroids and in the sections of murine fibrotic liver. RESULTS TGF-β-activated human hepatic and pancreatic stellate cells showed an increase of TLR5 expression. TLR5 knockdown blocked the activation of those stellate cells. Furthermore, TLR5 busted during murine liver fibrosis and co-localised with the inducible Collagen I. Flagellin suppressed TLR5, COL1A1 and ACTA2 expression after the administration of TGF-β. Instead, the antagonist of TLR5 did not block the effect of TGF-β. Wortmannin, a specific AKT inhibitor, induced TLR5 but not COL1A1 and ACTA2 transcript and protein level. CONCLUSION TGF-β-mediated activation of hepatic and pancreatic stellate cells requires the over-expression of TLR5. Instead, its autonomous signalling inhibits the activation of the stellate cells, thus prompting a signalling through different regulatory pathways.
Collapse
Affiliation(s)
- Pietro Di Fazio
- Department of Visceral Thoracic and Vascular Surgery, Philipps-Universität Marburg, Marburg, Germany
| | - Sophia Mielke
- Department of Visceral Thoracic and Vascular Surgery, Philipps-Universität Marburg, Marburg, Germany
| | - Isabell T Böhm
- Department of Visceral Thoracic and Vascular Surgery, Philipps-Universität Marburg, Marburg, Germany
| | - Malte Buchholz
- Department of Gastroenterology, Philipps-Universität Marburg, Marburg, Germany
| | - Sami Matrood
- Department of Visceral Thoracic and Vascular Surgery, Philipps-Universität Marburg, Marburg, Germany
| | - Detlef Schuppan
- Institute of Translational Immunology, Johannes Gutenberg Universitat Mainz, Mainz, Germany
- Division of Gastroenterology, Beth Israel Deaconess Medical Center, Boston, Massachusetts, USA
| | | |
Collapse
|
277
|
Yang W, Pan L, Cheng Y, Wu X, Huang S, Du J, Zhu H, Zhang M, Zhang Y. Amifostine attenuates bleomycin-induced pulmonary fibrosis in mice through inhibition of the PI3K/Akt/mTOR signaling pathway. Sci Rep 2023; 13:10485. [PMID: 37380638 DOI: 10.1038/s41598-023-34060-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2022] [Accepted: 04/24/2023] [Indexed: 06/30/2023] Open
Abstract
Amifostine is a normal cell protection agent, not only used in the adjuvant therapy of lung cancer, ovarian cancer, breast cancer, nasopharyngeal cancer, bone tumor, digestive tract tumor, blood system tumor and other cancers in order to reduce the toxicity of chemotherapy drugs, and recent studies have reported that the drug can also reduce lung tissue damage in patients with pulmonary fibrosis, but its mechanism of action is not yet fully understood. In this study, we explored the potential therapeutic effects and molecular mechanisms of AMI on bleomycin (BLM)-induced pulmonary fibrosis in mice. A mouse model of pulmonary fibrosis was established using BLM. We then assessed histopathological changes, inflammatory factors, oxidative indicators, apoptosis, epithelial-mesenchymal transition, extracellular matrix changes, and levels of phosphatidylinositol 3-kinase (PI3K)/Akt/mammalian target of rapamycin (mTOR) signaling pathway-related proteins in the BLM-treated mice to determine the effect of AMI treatment on these factors. BLM-treated mice had substantial lung inflammation and abnormal extracellular matrix deposition. Overall, treatment with AMI significantly improved BLM-induced lung injury and pulmonary fibrosis. More specifically, AMI alleviated BLM-induced oxidative stress, inflammation, alveolar cell apoptosis, epithelial-mesenchymal transition, and extracellular matrix deposition by regulating the PI3K/Akt/mTOR signaling pathway. This finding that AMI can alleviate pulmonary fibrosis in a mouse model by inhibiting activation of the PI3K/Akt/mTOR signaling pathway lays a foundation for potential future clinical application of this agent in patients with pulmonary fibrosis.
Collapse
Affiliation(s)
- Wenting Yang
- Department of Respiratory and Critical Care Medicine, The Affiliated Hospital of Guizhou Medical University, Guiyang, 550004, China
| | - Lin Pan
- Department of Respiratory and Critical Care Medicine, The Affiliated Hospital of Guizhou Medical University, Guiyang, 550004, China
| | - Yiju Cheng
- Department of Respiratory and Critical Care Medicine, The First People's Hospital of Guiyang, Guiyang, 550004, China.
- Guizhou Medical University, Guiyang, 550004, China.
| | - Xiao Wu
- Department of Respiratory and Critical Care Medicine, The Affiliated Hospital of Guizhou Medical University, Guiyang, 550004, China
| | - Songsong Huang
- Department of Pathology, The Affiliated Hospital of Guizhou Medical University, Guiyang, 550004, China
| | - Juan Du
- Department of Respiratory and Critical Care Medicine, The Affiliated Hospital of Guizhou Medical University, Guiyang, 550004, China
| | - Honglan Zhu
- Department of Respiratory and Critical Care Medicine, The Affiliated Hospital of Guizhou Medical University, Guiyang, 550004, China
| | - Menglin Zhang
- Department of Respiratory and Critical Care Medicine, The Affiliated Hospital of Guizhou Medical University, Guiyang, 550004, China
| | - Yuquan Zhang
- Department of Respiratory and Critical Care Medicine, The Affiliated Hospital of Guizhou Medical University, Guiyang, 550004, China
| |
Collapse
|
278
|
Zhao XH, Zhao P, Deng Z, Yang T, Qi YX, An LY, Sun DL, He HY. Integrative analysis reveals marker genes for intestinal mucosa barrier repairing in clinical patients. iScience 2023; 26:106831. [PMID: 37250791 PMCID: PMC10212979 DOI: 10.1016/j.isci.2023.106831] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2022] [Revised: 03/21/2023] [Accepted: 05/03/2023] [Indexed: 05/31/2023] Open
Abstract
This study aims to identify biomarkers of intestinal repair and provide potential therapeutic clues for improving functional recovery and prognostic performance after intestinal inflammation or injury. Here, we conducted a large-scale screening of multiple transcriptomic and scRNA-seq datasets of patients with inflammatory bowel disease (IBD), and identified 10 marker genes that potentially contribute to intestinal barrier repairing: AQP8, SULT1A1, HSD17B2, PADI2, SLC26A2, SELENBP1, FAM162A, TNNC2, ACADS, and TST. Analysis of a published scRNA-seq dataset revealed that expression of these healing markers were specific to absorptive cell types in intestinal epithelium. Furthermore, we conducted a clinical study where 11 patients underwent ileum resection demonstrating that upregulation of post-operative AQP8 and SULT1A1 expression were associated with improved recovery of bowel functions after surgery-induced intestinal injury, making them confident biomarkers of intestinal healing as well as potential prognostic markers and therapeutic targets for patients with impaired intestinal barrier functions.
Collapse
Affiliation(s)
- Xiao-Hu Zhao
- Department of Gastrointestinal Surgery, Second Affiliated Hospital of Kunming Medical University / Second Faculty of Clinical Medicine, Kunming Medical University, Kunming 650101, China
| | - Peinan Zhao
- Department of Medicine (Alfred Hospital), Central Clinical School, Monash University, 99 Commercial Rd, Melbourne, VIC 3004, Australia
| | - Zihao Deng
- Department of Medicine (Alfred Hospital), Central Clinical School, Monash University, 99 Commercial Rd, Melbourne, VIC 3004, Australia
| | - Ting Yang
- Department of Gastrointestinal Surgery, Second Affiliated Hospital of Kunming Medical University / Second Faculty of Clinical Medicine, Kunming Medical University, Kunming 650101, China
| | - Yu-Xing Qi
- Department of Gastrointestinal Surgery, Second Affiliated Hospital of Kunming Medical University / Second Faculty of Clinical Medicine, Kunming Medical University, Kunming 650101, China
| | - Li-Ya An
- Department of Gastrointestinal Surgery, Second Affiliated Hospital of Kunming Medical University / Second Faculty of Clinical Medicine, Kunming Medical University, Kunming 650101, China
| | - Da-Li Sun
- Department of Gastrointestinal Surgery, Second Affiliated Hospital of Kunming Medical University / Second Faculty of Clinical Medicine, Kunming Medical University, Kunming 650101, China
| | - Hai-Yu He
- Department of Gastroenterology, Second Affiliated Hospital of Kunming Medical University / Second Faculty of Clinical Medicine, Kunming Medical University, Kunming 650101, China
| |
Collapse
|
279
|
Roman J. Fibroblasts-Warriors at the Intersection of Wound Healing and Disrepair. Biomolecules 2023; 13:945. [PMID: 37371525 DOI: 10.3390/biom13060945] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2023] [Revised: 05/07/2023] [Accepted: 05/17/2023] [Indexed: 06/29/2023] Open
Abstract
Wound healing is triggered by inflammation elicited after tissue injury. Mesenchymal cells, specifically fibroblasts, accumulate in the injured tissues, where they engage in tissue repair through the expression and assembly of extracellular matrices that provide a scaffold for cell adhesion, the re-epithelialization of tissues, the production of soluble bioactive mediators that promote cellular recruitment and differentiation, and the regulation of immune responses. If appropriately deployed, these processes promote adaptive repair, resulting in the preservation of the tissue structure and function. Conversely, the dysregulation of these processes leads to maladaptive repair or disrepair, which causes tissue destruction and a loss of organ function. Thus, fibroblasts not only serve as structural cells that maintain tissue integrity, but are key effector cells in the process of wound healing. The review will discuss the general concepts about the origins and heterogeneity of this cell population and highlight the specific fibroblast functions disrupted in human disease. Finally, the review will explore the role of fibroblasts in tissue disrepair, with special attention to the lung, the role of aging, and how alterations in the fibroblast phenotype underpin disorders characterized by pulmonary fibrosis.
Collapse
Affiliation(s)
- Jesse Roman
- Department of Medicine, Division of Pulmonary, Allergy and Critical Care and The Jane & Leonard Korman Respiratory Institute, Thomas Jefferson University, Philadelphia, PA 19107, USA
| |
Collapse
|
280
|
Chang C, Guo W, Yu X, Guo C, Zhou N, Guo X, Huang RL, Li Q, Zhu Y. Engineered M13 phage as a novel therapeutic bionanomaterial for clinical applications: From tissue regeneration to cancer therapy. Mater Today Bio 2023; 20:100612. [PMID: 37063776 PMCID: PMC10102448 DOI: 10.1016/j.mtbio.2023.100612] [Citation(s) in RCA: 24] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2023] [Revised: 03/12/2023] [Accepted: 03/15/2023] [Indexed: 04/03/2023] Open
Abstract
Bacteriophages (phages) are nanostructured viruses with highly selective antibacterial properties that have gained attention beyond eliminating bacteria. Specifically, M13 phages are filamentous phages that have recently been studied in various aspects of nanomedicine due to their biological advantages and more compliant engineering capabilities over other phages. Having nanofiber-like morphology, M13 phages can reach varied target sites and self-assemble into multidimensional scaffolds in a relatively safe and stable way. In addition, genetic modification of the coat proteins enables specific display of peptides and antibodies on the phages, allowing for precise and individualized medicine. M13 phages have also been subjected to novel engineering approaches, including phage-based bionanomaterial engineering and phage-directed nanomaterial combinations that enhance the bionanomaterial properties of M13 phages. In view of these features, researchers have been able to utilize M13 phages for therapeutic applications such as drug delivery, biodetection, tissue regeneration, and targeted cancer therapy. In particular, M13 phages have been utilized as a novel bionanomaterial for precisely mimicking natural tissue environment in order to overcome the shortage in tissue and organ donors. Hence, in this review, we address the recent studies and advances of using M13 phages in the field of nanomedicine as therapeutic agents based upon their characteristics as novel bionanomaterial with biomolecules displayed. This paper also emphasizes the novel engineering approach that enhances M13 phage's bionanomaterial capabilities. Current limitations and future approaches are also discussed to provide insight in further progress for M13 phage-based clinical applications.
Collapse
Affiliation(s)
- Cheng Chang
- School of Global Health, Chinese Centre for Tropical Diseases Research, Shanghai Jiao Tong University School of Medicine, One Health Center, Shanghai Jiao Tong University-The University of Edinburgh, Shanghai, 200025, China
| | - Wennan Guo
- School of Global Health, Chinese Centre for Tropical Diseases Research, Shanghai Jiao Tong University School of Medicine, One Health Center, Shanghai Jiao Tong University-The University of Edinburgh, Shanghai, 200025, China
| | - Xinbo Yu
- Second Dental Center, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 201999, China
| | - Chaoyi Guo
- School of Global Health, Chinese Centre for Tropical Diseases Research, Shanghai Jiao Tong University School of Medicine, One Health Center, Shanghai Jiao Tong University-The University of Edinburgh, Shanghai, 200025, China
| | - Nan Zhou
- School of Global Health, Chinese Centre for Tropical Diseases Research, Shanghai Jiao Tong University School of Medicine, One Health Center, Shanghai Jiao Tong University-The University of Edinburgh, Shanghai, 200025, China
| | - Xiaokui Guo
- School of Global Health, Chinese Centre for Tropical Diseases Research, Shanghai Jiao Tong University School of Medicine, One Health Center, Shanghai Jiao Tong University-The University of Edinburgh, Shanghai, 200025, China
| | - Ru-Lin Huang
- Department of Plastic and Reconstructive Surgery, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200011, China
- Corresponding author.
| | - Qingtian Li
- Department of Laboratory Medicine, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, China
- Corresponding author.
| | - Yongzhang Zhu
- School of Global Health, Chinese Centre for Tropical Diseases Research, Shanghai Jiao Tong University School of Medicine, One Health Center, Shanghai Jiao Tong University-The University of Edinburgh, Shanghai, 200025, China
- Corresponding author.
| |
Collapse
|
281
|
Hu KH, Kuhn NF, Courau T, Tsui J, Samad B, Ha P, Kratz JR, Combes AJ, Krummel MF. Transcriptional space-time mapping identifies concerted immune and stromal cell patterns and gene programs in wound healing and cancer. Cell Stem Cell 2023; 30:885-903.e10. [PMID: 37267918 PMCID: PMC10843988 DOI: 10.1016/j.stem.2023.05.001] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2022] [Revised: 03/13/2023] [Accepted: 05/02/2023] [Indexed: 06/04/2023]
Abstract
Tissue repair responses in metazoans are highly coordinated by different cell types over space and time. However, comprehensive single-cell-based characterization covering this coordination is lacking. Here, we captured transcriptional states of single cells over space and time during skin wound closure, revealing choreographed gene-expression profiles. We identified shared space-time patterns of cellular and gene program enrichment, which we call multicellular "movements" spanning multiple cell types. We validated some of the discovered space-time movements using large-volume imaging of cleared wounds and demonstrated the value of this analysis to predict "sender" and "receiver" gene programs in macrophages and fibroblasts. Finally, we tested the hypothesis that tumors are like "wounds that never heal" and found conserved wound healing movements in mouse melanoma and colorectal tumor models, as well as human tumor samples, revealing fundamental multicellular units of tissue biology for integrative studies.
Collapse
Affiliation(s)
- Kenneth H Hu
- Department of Pathology, University of California, San Francisco, San Francisco, CA 94143, USA; ImmunoX Initiative, University of California, San Francisco, San Francisco, CA 94143, USA.
| | - Nicholas F Kuhn
- Department of Pathology, University of California, San Francisco, San Francisco, CA 94143, USA; ImmunoX Initiative, University of California, San Francisco, San Francisco, CA 94143, USA
| | - Tristan Courau
- Department of Pathology, University of California, San Francisco, San Francisco, CA 94143, USA; ImmunoX Initiative, University of California, San Francisco, San Francisco, CA 94143, USA; UCSF CoLabs, University of California, San Francisco, San Francisco, CA 94143, USA
| | - Jessica Tsui
- ImmunoX Initiative, University of California, San Francisco, San Francisco, CA 94143, USA; UCSF CoLabs, University of California, San Francisco, San Francisco, CA 94143, USA; Department of Otolaryngology Head and Neck Surgery, University of California, San Francisco, San Francisco, CA 94143, USA
| | - Bushra Samad
- Department of Pathology, University of California, San Francisco, San Francisco, CA 94143, USA; ImmunoX Initiative, University of California, San Francisco, San Francisco, CA 94143, USA; UCSF CoLabs, University of California, San Francisco, San Francisco, CA 94143, USA
| | - Patrick Ha
- Department of Otolaryngology Head and Neck Surgery, University of California, San Francisco, San Francisco, CA 94143, USA
| | - Johannes R Kratz
- ImmunoX Initiative, University of California, San Francisco, San Francisco, CA 94143, USA; Department of Surgery, University of California, San Francisco, San Francisco, CA 94143, USA
| | - Alexis J Combes
- Department of Pathology, University of California, San Francisco, San Francisco, CA 94143, USA; ImmunoX Initiative, University of California, San Francisco, San Francisco, CA 94143, USA; UCSF CoLabs, University of California, San Francisco, San Francisco, CA 94143, USA
| | - Matthew F Krummel
- Department of Pathology, University of California, San Francisco, San Francisco, CA 94143, USA; ImmunoX Initiative, University of California, San Francisco, San Francisco, CA 94143, USA.
| |
Collapse
|
282
|
Okamura DM, Nguyen ED, Collins SJ, Yoon K, Gere JB, Weiser-Evans MCM, Beier DR, Majesky MW. Mammalian organ regeneration in spiny mice. J Muscle Res Cell Motil 2023; 44:39-52. [PMID: 36131170 DOI: 10.1007/s10974-022-09631-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2022] [Accepted: 08/30/2022] [Indexed: 11/26/2022]
Abstract
Fibrosis-driven solid organ failure is a major world-wide health burden with few therapeutic options. Spiny mice (genus: Acomys) are terrestrial mammals that regenerate severe skin wounds without fibrotic scars to evade predators. Recent studies have shown that spiny mice also regenerate acute ischemic and traumatic injuries to kidney, heart, spinal cord, and skeletal muscle. A common feature of this evolved wound healing response is a lack of formation of fibrotic scar tissue that degrades organ function, inhibits regeneration, and leads to organ failure. Complex tissue regeneration is an extremely rare property among mammalian species. In this article, we discuss the evidence that Acomys represents an emerging model organism that offers a unique opportunity for the biomedical community to investigate and clinically translate molecular mechanisms of scarless wound healing and regeneration of organ function in a mammalian species.
Collapse
Affiliation(s)
- Daryl M Okamura
- Center for Developmental Biology & Regenerative Medicine, Seattle Children's Research Institute, 1900 Ninth Avenue, M/S C9S-5, Seattle, WA, 98101, USA
- Department of Pediatrics, University of Washington, Seattle, WA, 98195, USA
| | - Elizabeth D Nguyen
- Center for Developmental Biology & Regenerative Medicine, Seattle Children's Research Institute, 1900 Ninth Avenue, M/S C9S-5, Seattle, WA, 98101, USA
- Department of Pediatrics, University of Washington, Seattle, WA, 98195, USA
| | - Sarah J Collins
- Center for Developmental Biology & Regenerative Medicine, Seattle Children's Research Institute, 1900 Ninth Avenue, M/S C9S-5, Seattle, WA, 98101, USA
| | - Kevin Yoon
- Center for Developmental Biology & Regenerative Medicine, Seattle Children's Research Institute, 1900 Ninth Avenue, M/S C9S-5, Seattle, WA, 98101, USA
| | - Joshua B Gere
- Center for Developmental Biology & Regenerative Medicine, Seattle Children's Research Institute, 1900 Ninth Avenue, M/S C9S-5, Seattle, WA, 98101, USA
| | - Mary C M Weiser-Evans
- Department of Medicine, Division of Renal Diseases & Hypertension, University of Colorado Anschutz Medical Campus, Aurora, CO, 80045, USA
| | - David R Beier
- Center for Developmental Biology & Regenerative Medicine, Seattle Children's Research Institute, 1900 Ninth Avenue, M/S C9S-5, Seattle, WA, 98101, USA
- Department of Pediatrics, University of Washington, Seattle, WA, 98195, USA
| | - Mark W Majesky
- Center for Developmental Biology & Regenerative Medicine, Seattle Children's Research Institute, 1900 Ninth Avenue, M/S C9S-5, Seattle, WA, 98101, USA.
- Department of Pediatrics, University of Washington, Seattle, WA, 98195, USA.
- Department of Laboratory Medicine & Pathology, University of Washington, Seattle, WA, 98195, USA.
- Institute for Stem Cell & Regenerative Medicine, University of Washington, Seattle, WA, 98195, USA.
| |
Collapse
|
283
|
Cartwright BM, Fox SJ, Underdown MJ, Clark WA, Molnar JA. ARAG, an Antioxidant-Rich Gel, Shows Superiority to Mepilex Ag in the Treatment of Deep Partial Thickness Burns without Sacrificing Antimicrobial Efficiency. Antioxidants (Basel) 2023; 12:1176. [PMID: 37371906 DOI: 10.3390/antiox12061176] [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: 04/15/2023] [Revised: 05/23/2023] [Accepted: 05/26/2023] [Indexed: 06/29/2023] Open
Abstract
Current treatments for deep tissue burns are limited, and most serve only to enhance hydration or prevent bacterial growth. This leaves burn healing dependent on slow natural processes to debride the wound and reestablish the epidermal and dermal layers of the skin. Infections are well known to destabilize this process through a variety of mechanisms, most notably through increased inflammation and the resulting oxidative stress. In this study, we show that ARAG (an antioxidant-rich antimicrobial gel) can suppress the growth of multiple bacteria commonly found to infect burns (Klebsiella pneumoniae, Proteus vulgaris, Pseudomonas aeruginosa, and Staphylococcus aureus). This inhibition is comparable to that conferred by silver ion release from burn dressings such as Mepilex-Ag. We further show, using a porcine model for deep partial-thickness burns, that ARAG allows for enhanced wound healing over Mepilex-Ag, the current standard of care. Histological findings indicate this is likely due to increased wound debridement and dampening of late inflammatory processes, leading to more balanced physiologic healing. Taken together, these findings show promise for ARAG as a superior alternative to the current standard of care.
Collapse
Affiliation(s)
- Brian Michael Cartwright
- ETSU Quillen College of Medicine, East Tennessee State University, Johnson City, TN 37614, USA
- Department of Rehabilitative Sciences, College of Clinical and Rehabilitative Health Sciences, East Tennessee State University, Johnson City, TN 37614, USA
| | - Sean James Fox
- Department of Health Sciences, College of Public Health, East Tennessee State University, Johnson City, TN 37614, USA
| | - Mary Jane Underdown
- Department of Rehabilitative Sciences, College of Clinical and Rehabilitative Health Sciences, East Tennessee State University, Johnson City, TN 37614, USA
| | - William Andrew Clark
- Department of Rehabilitative Sciences, College of Clinical and Rehabilitative Health Sciences, East Tennessee State University, Johnson City, TN 37614, USA
| | - Joseph Andrew Molnar
- Department of Plastic and Reconstructive Surgery, Wake Forest University School of Medicine, Winston-Salem, NC 27101, USA
- Wake Forest Institute for Regenerative Medicine, Wake Forest University School of Medicine, Winston-Salem, NC 27101, USA
| |
Collapse
|
284
|
Han C, Leonardo TR, Romana-Souza B, Shi J, Keiser S, Yuan H, Altakriti M, Ranzer MJ, Ferri-Borgogno S, Mok SC, Koh TJ, Hong SJ, Chen L, DiPietro LA. Microfibril-associated protein 5 and the regulation of skin scar formation. Sci Rep 2023; 13:8728. [PMID: 37253753 PMCID: PMC10229580 DOI: 10.1038/s41598-023-35558-x] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2023] [Accepted: 05/20/2023] [Indexed: 06/01/2023] Open
Abstract
Many factors regulate scar formation, which yields a modified extracellular matrix (ECM). Among ECM components, microfibril-associated proteins have been minimally explored in the context of skin wound repair. Microfibril-associated protein 5 (MFAP5), a small 25 kD serine and threonine rich microfibril-associated protein, influences microfibril function and modulates major extracellular signaling pathways. Though known to be associated with fibrosis and angiogenesis in certain pathologies, MFAP5's role in wound healing is unknown. Using a murine model of skin wound repair, we found that MFAP5 is significantly expressed during the proliferative and remodeling phases of healing. Analysis of existing single-cell RNA-sequencing data from mouse skin wounds identified two fibroblast subpopulations as the main expressors of MFAP5 during wound healing. Furthermore, neutralization of MFAP5 in healing mouse wounds decreased collagen deposition and refined angiogenesis without altering wound closure. In vitro, recombinant MFAP5 significantly enhanced dermal fibroblast migration, collagen contractility, and expression of pro-fibrotic genes. Additionally, TGF-ß1 increased MFAP5 expression and production in dermal fibroblasts. Our findings suggest that MFAP5 regulates fibroblast function and influences scar formation in healing wounds. Our work demonstrates a previously undescribed role for MFAP5 and suggests that microfibril-associated proteins may be significant modulators of wound healing outcomes and scarring.
Collapse
Affiliation(s)
- Chen Han
- Center for Wound Healing and Tissue Regeneration, University of Illinois Chicago, Chicago, IL, USA
| | - Trevor R Leonardo
- Department of Microbiology and Immunology, College of Medicine, University of Illinois Chicago, Chicago, IL, USA
| | - Bruna Romana-Souza
- Department of Histology and Embryology, Rio de Janeiro State University, Rio de Janeiro, RJ, Brazil
| | - Junhe Shi
- NMPA Key Laboratory for Clinical Research and Evaluation of Traditional Chinese Medicine, Xiyuan Hospital, China Academy of Chinese Medical Sciences, Beijing, China
| | - Shalyn Keiser
- Center for Wound Healing and Tissue Regeneration, University of Illinois Chicago, Chicago, IL, USA
| | - Heidi Yuan
- Center for Wound Healing and Tissue Regeneration, University of Illinois Chicago, Chicago, IL, USA
| | - Mohamad Altakriti
- Center for Wound Healing and Tissue Regeneration, University of Illinois Chicago, Chicago, IL, USA
| | - Matthew J Ranzer
- Department of Surgery, University of Illinois Chicago, Chicago, IL, USA
| | - Sammy Ferri-Borgogno
- Department of Gynecologic Oncology and Reproductive Medicine, University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Samuel C Mok
- Department of Gynecologic Oncology and Reproductive Medicine, University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Timothy J Koh
- Center for Wound Healing and Tissue Regeneration, University of Illinois Chicago, Chicago, IL, USA
- Department of Kinesiology and Nutrition, University of Illinois Chicago, Chicago, IL, USA
| | - Seok Jong Hong
- Department of Surgery, Northwestern University-Feinberg School of Medicine, Chicago, IL, USA
| | - Lin Chen
- Center for Wound Healing and Tissue Regeneration, University of Illinois Chicago, Chicago, IL, USA.
| | - Luisa A DiPietro
- Center for Wound Healing and Tissue Regeneration, University of Illinois Chicago, Chicago, IL, USA.
| |
Collapse
|
285
|
Maddali P, Ambesi A, McKeown-Longo PJ. Induction of pro-inflammatory genes by fibronectin DAMPs in three fibroblast cell lines: Role of TAK1 and MAP kinases. PLoS One 2023; 18:e0286390. [PMID: 37228128 DOI: 10.1371/journal.pone.0286390] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2023] [Accepted: 05/13/2023] [Indexed: 05/27/2023] Open
Abstract
Changes in the organization and structure of the fibronectin matrix are believed to contribute to dysregulated wound healing and subsequent tissue inflammation and tissue fibrosis. These changes include an increase in the EDA isoform of fibronectin as well as the mechanical unfolding of fibronectin type III domains. In previous studies using embryonic foreskin fibroblasts, we have shown that fibronectin's EDA domain (FnEDA) and the partially unfolded first Type III domain (FnIII-1c) function as Damage Associated Molecular Pattern (DAMP) molecules to stimulate the induction of inflammatory cytokines by serving as agonists for Toll-Like Receptor-4 (TLR4). However, the role of signaling molecules downstream of TLR-4 such as TGF-β Activated Kinase 1 (TAK1) and Mitogen activated protein kinases (MAPK) in regulating the expression of fibronectin DAMP induced inflammatory genes in specific cell types is not known. In the current study, we evaluate the molecular steps regulating the fibronectin driven induction of inflammatory genes in three human fibroblast cell lines: embryonic foreskin, adult dermal, and adult kidney. The fibronectin derived DAMPs each induce the phosphorylation and activation of TAK1 which results in the activation of two downstream signaling arms, IKK/NF-κB and MAPK. Using the specific inhibitor 5Z-(7)-Oxozeanol as well as siRNA, we show TAK1 to be a crucial signaling mediator in the release of cytokines in response to fibronectin DAMPs in all three cell types. Finally, we show that FnEDA and FnIII-1c induce several pro-inflammatory cytokines whose expression is dependent on both TAK1 and JNK MAPK and highlight cell-type specific differences in the gene-expression profiles of the fibroblast cell-lines.
Collapse
Affiliation(s)
- Pranav Maddali
- Department of Regenerative & Cancer Cell Biology, Albany Medical College, Albany, New York, United States of America
| | - Anthony Ambesi
- Department of Regenerative & Cancer Cell Biology, Albany Medical College, Albany, New York, United States of America
| | - Paula J McKeown-Longo
- Department of Regenerative & Cancer Cell Biology, Albany Medical College, Albany, New York, United States of America
| |
Collapse
|
286
|
Graca FA, Stephan A, Minden-Birkenmaier BA, Shirinifard A, Wang YD, Demontis F, Labelle M. Platelet-derived chemokines promote skeletal muscle regeneration by guiding neutrophil recruitment to injured muscles. Nat Commun 2023; 14:2900. [PMID: 37217480 DOI: 10.1038/s41467-023-38624-0] [Citation(s) in RCA: 16] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2022] [Accepted: 05/09/2023] [Indexed: 05/24/2023] Open
Abstract
Skeletal muscle regeneration involves coordinated interactions between different cell types. Injection of platelet-rich plasma is circumstantially considered an aid to muscle repair but whether platelets promote regeneration beyond their role in hemostasis remains unexplored. Here, we find that signaling via platelet-released chemokines is an early event necessary for muscle repair in mice. Platelet depletion reduces the levels of the platelet-secreted neutrophil chemoattractants CXCL5 and CXCL7/PPBP. Consequently, early-phase neutrophil infiltration to injured muscles is impaired whereas later inflammation is exacerbated. Consistent with this model, neutrophil infiltration to injured muscles is compromised in male mice with Cxcl7-knockout platelets. Moreover, neo-angiogenesis and the re-establishment of myofiber size and muscle strength occurs optimally in control mice post-injury but not in Cxcl7ko mice and in neutrophil-depleted mice. Altogether, these findings indicate that platelet-secreted CXCL7 promotes regeneration by recruiting neutrophils to injured muscles, and that this signaling axis could be utilized therapeutically to boost muscle regeneration.
Collapse
Affiliation(s)
- Flavia A Graca
- Department of Developmental Neurobiology, St. Jude Children's Research Hospital, Memphis, TN, 38105, USA
| | - Anna Stephan
- Department of Developmental Neurobiology, St. Jude Children's Research Hospital, Memphis, TN, 38105, USA
| | - Benjamin A Minden-Birkenmaier
- Department of Developmental Neurobiology, St. Jude Children's Research Hospital, Memphis, TN, 38105, USA
- Department of Oncology, Division of Molecular Oncology, St. Jude Children's Research Hospital, Memphis, TN, 38105, USA
| | - Abbas Shirinifard
- Department of Developmental Neurobiology, St. Jude Children's Research Hospital, Memphis, TN, 38105, USA
| | - Yong-Dong Wang
- Department of Cell and Molecular Biology, St. Jude Children's Research Hospital, Memphis, TN, 38105, USA
| | - Fabio Demontis
- Department of Developmental Neurobiology, St. Jude Children's Research Hospital, Memphis, TN, 38105, USA.
| | - Myriam Labelle
- Department of Developmental Neurobiology, St. Jude Children's Research Hospital, Memphis, TN, 38105, USA.
- Department of Oncology, Division of Molecular Oncology, St. Jude Children's Research Hospital, Memphis, TN, 38105, USA.
| |
Collapse
|
287
|
Rizzo MG, Best TM, Huard J, Philippon M, Hornicek F, Duan Z, Griswold AJ, Kaplan LD, Hare JM, Kouroupis D. Therapeutic Perspectives for Inflammation and Senescence in Osteoarthritis Using Mesenchymal Stem Cells, Mesenchymal Stem Cell-Derived Extracellular Vesicles and Senolytic Agents. Cells 2023; 12:1421. [PMID: 37408255 PMCID: PMC10217382 DOI: 10.3390/cells12101421] [Citation(s) in RCA: 23] [Impact Index Per Article: 11.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2023] [Revised: 05/08/2023] [Accepted: 05/13/2023] [Indexed: 07/07/2023] Open
Abstract
Osteoarthritis (OA) is the most common cause of disability worldwide among the elderly. Alarmingly, the incidence of OA in individuals less than 40 years of age is rising, likely due to the increase in obesity and post-traumatic osteoarthritis (PTOA). In recent years, due to a better understanding of the underlying pathophysiology of OA, several potential therapeutic approaches targeting specific molecular pathways have been identified. In particular, the role of inflammation and the immune system has been increasingly recognized as important in a variety of musculoskeletal diseases, including OA. Similarly, higher levels of host cellular senescence, characterized by cessation of cell division and the secretion of a senescence-associated secretory phenotype (SASP) within the local tissue microenvironments, have also been linked to OA and its progression. New advances in the field, including stem cell therapies and senolytics, are emerging with the goal of slowing disease progression. Mesenchymal stem/stromal cells (MSCs) are a subset of multipotent adult stem cells that have demonstrated the potential to modulate unchecked inflammation, reverse fibrosis, attenuate pain, and potentially treat patients with OA. Numerous studies have demonstrated the potential of MSC extracellular vesicles (EVs) as cell-free treatments that comply with FDA regulations. EVs, including exosomes and microvesicles, are released by numerous cell types and are increasingly recognized as playing a critical role in cell-cell communication in age-related diseases, including OA. Treatment strategies for OA are being developed that target senescent cells and the paracrine and autocrine secretions of SASP. This article highlights the encouraging potential for MSC or MSC-derived products alone or in combination with senolytics to control patient symptoms and potentially mitigate the progression of OA. We will also explore the application of genomic principles to the study of OA and the potential for the discovery of OA phenotypes that can motivate more precise patient-driven treatments.
Collapse
Affiliation(s)
- Michael G. Rizzo
- Department of Orthopedics, UHealth Sports Medicine Institute, University of Miami Miller School of Medicine, Miami, FL 33146, USA; (M.G.R.); (T.M.B.)
| | - Thomas M. Best
- Department of Orthopedics, UHealth Sports Medicine Institute, University of Miami Miller School of Medicine, Miami, FL 33146, USA; (M.G.R.); (T.M.B.)
| | - Johnny Huard
- Center for Regenerative and Personalized Medicine (CRPM), Steadman Philippon Research Institute, Vail, CO 81657, USA (M.P.)
| | - Marc Philippon
- Center for Regenerative and Personalized Medicine (CRPM), Steadman Philippon Research Institute, Vail, CO 81657, USA (M.P.)
| | - Francis Hornicek
- Department of Orthopedics, Sarcoma Biology Laboratory, Sylvester Comprehensive Cancer Center, University of Miami Miller School of Medicine, Miami, FL 33136, USA; (F.H.); (Z.D.)
| | - Zhenfeng Duan
- Department of Orthopedics, Sarcoma Biology Laboratory, Sylvester Comprehensive Cancer Center, University of Miami Miller School of Medicine, Miami, FL 33136, USA; (F.H.); (Z.D.)
| | - Anthony J. Griswold
- John P. Hussman Institute for Human Genomics, University of Miami Miller School of Medicine, Miami, FL 33136, USA;
| | - Lee D. Kaplan
- Department of Orthopedics, UHealth Sports Medicine Institute, University of Miami Miller School of Medicine, Miami, FL 33146, USA; (M.G.R.); (T.M.B.)
| | - Joshua M. Hare
- Interdisciplinary Stem Cell Institute, University of Miami Miller School of Medicine, Miami, FL 33101, USA
| | - Dimitrios Kouroupis
- Department of Orthopedics, UHealth Sports Medicine Institute, University of Miami Miller School of Medicine, Miami, FL 33146, USA; (M.G.R.); (T.M.B.)
- Diabetes Research Institute, Cell Transplant Center, University of Miami Miller School of Medicine, Miami, FL 33136, USA
| |
Collapse
|
288
|
Zheng Y, Dong X, Wang X, Wang J, Chen S, He Y, An J, He L, Zhang Y. Exosomes Derived from Adipose Tissue-Derived Mesenchymal Stromal Cells Prevent Medication-Related Osteonecrosis of the Jaw through IL-1RA. Int J Mol Sci 2023; 24:ijms24108694. [PMID: 37240036 DOI: 10.3390/ijms24108694] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2023] [Revised: 03/20/2023] [Accepted: 03/21/2023] [Indexed: 05/28/2023] Open
Abstract
Medication-related osteonecrosis of the jaw (MRONJ) is a severe disease with unclear pathogenesis. Adipose tissue-derived mesenchymal stromal cells (MSC(AT)s) serve as a special source for cell therapy. Herein, we explored whether exosomes (Exo) derived from MSC(AT)s promote primary gingival wound healing and prevent MRONJ. An MRONJ mice model was constructed using zoledronate (Zol) administration and tooth extraction. Exosomes were collected from the conditioned medium (CM) of MSC(AT)s (MSC(AT)s-Exo) and locally administered into the tooth sockets. Interleukin-1 receptor antagonist (IL-1RA)-siRNA was used to knock down the expression of IL-1RA in MSC(AT)s-Exo. Clinical observations, micro-computed tomography (microCT), and histological analysis were used to evaluate the therapeutic effects in vivo. In addition, the effect of exosomes on the biological behavior of human gingival fibroblasts (HGFs) was evaluated in vitro. MSC(AT)s-Exo accelerated primary gingival wound healing and bone regeneration in tooth sockets and prevented MRONJ. Moreover, MSC(AT)s-Exo increased IL-1RA expression and decreased interleukin-1 beta (IL-1β) and tumor necrosis factor-α (TNF-α) expression in the gingival tissue. The sequent rescue assay showed that the effects of preventing MRONJ in vivo and improving the migration and collagen synthesis abilities of zoledronate-affected HGFs in vitro were partially impaired in the IL-1RA-deficient exosome group. Our results indicated that MSC(AT)s-Exo might prevent the onset of MRONJ via an IL-1RA-mediated anti-inflammatory effect in the gingiva wound and improve the migration and collagen synthesis abilities of HGFs.
Collapse
Affiliation(s)
- Yi Zheng
- Department of Oral and Maxillofacial Surgery, Peking University School and Hospital of Stomatology, Beijing 100081, China
| | - Xian Dong
- Department of Oral and Maxillofacial Surgery, Peking University School and Hospital of Stomatology, Beijing 100081, China
- First Clinical Division, Peking University School and Hospital of Stomatology, Beijing 100081, China
| | - Xinyu Wang
- Department of Oral and Maxillofacial Surgery, Peking University School and Hospital of Stomatology, Beijing 100081, China
| | - Jie Wang
- Department of Oral and Maxillofacial Surgery, Peking University School and Hospital of Stomatology, Beijing 100081, China
| | - Shuo Chen
- Department of Oral and Maxillofacial Surgery, Peking University School and Hospital of Stomatology, Beijing 100081, China
| | - Yang He
- Department of Oral and Maxillofacial Surgery, Peking University School and Hospital of Stomatology, Beijing 100081, China
| | - Jingang An
- Department of Oral and Maxillofacial Surgery, Peking University School and Hospital of Stomatology, Beijing 100081, China
| | - Linhai He
- Department of Oral and Maxillofacial Surgery, Peking University School and Hospital of Stomatology, Beijing 100081, China
- First Clinical Division, Peking University School and Hospital of Stomatology, Beijing 100081, China
| | - Yi Zhang
- Department of Oral and Maxillofacial Surgery, Peking University School and Hospital of Stomatology, Beijing 100081, China
| |
Collapse
|
289
|
Hou J, Chen J, Fan J, Tang Z, Zhou W, Lin H. Inhibition of NF-κB Signaling-Mediated Crosstalk Between Macrophages and Preosteoblasts by Metformin Alleviates Trauma-Induced Heterotopic Ossification. Inflammation 2023:10.1007/s10753-023-01817-2. [PMID: 37115368 DOI: 10.1007/s10753-023-01817-2] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2023] [Revised: 04/04/2023] [Accepted: 04/05/2023] [Indexed: 04/29/2023]
Abstract
Heterotopic ossification (HO) is a pathological condition that occurs in soft tissues following severe trauma. The exact pathogenesis of HO remains unclear. Studies have shown that inflammation predisposes patients to the development of HO and triggers ectopic bone formation. Macrophages are crucial mediators of inflammation and are involved in HO development. The present study investigated the inhibitory effect and underlying mechanism of metformin on macrophage infiltration and traumatic HO in mice. Our results found that abundant levels of macrophages were recruited to the injury site during early HO progression and that early administration of metformin prevented traumatic HO in mice. Furthermore, we found that metformin attenuated macrophage infiltration and the NF-κB signaling pathway in injured tissue. The monocyte-to-macrophage transition in vitro was suppressed by metformin and this event was mediated by AMPK. Finally, we showed that inflammatory mediator's regulation by macrophages targeted preosteoblasts, leading to elevated BMP signaling, and osteogenic differentiation and driving HO formation, and this effect was blocked after the activation of AMPK in macrophages. Collectively, our study suggests that metformin prevents traumatic HO by inhibiting of NF-κB signaling in macrophages and subsequently attenuating BMP signaling and osteogenic differentiation in preosteoblasts. Therefore, metformin may serve as a therapeutic drug for traumatic HO by targeting NF-κB signaling in macrophages.
Collapse
Affiliation(s)
- Jia Hou
- Department of Pathophysiology, School of Basic Medical Sciences, Nanchang University, Nanchang, 330006, Jiangxi, China
| | - Jie Chen
- Department of Pathophysiology, School of Basic Medical Sciences, Nanchang University, Nanchang, 330006, Jiangxi, China
| | - Jingjing Fan
- Department of Pathophysiology, School of Basic Medical Sciences, Nanchang University, Nanchang, 330006, Jiangxi, China
| | - Zhimin Tang
- Department of Pathophysiology, School of Basic Medical Sciences, Nanchang University, Nanchang, 330006, Jiangxi, China
| | - Wenwen Zhou
- Department of Pathophysiology, School of Basic Medical Sciences, Nanchang University, Nanchang, 330006, Jiangxi, China
| | - Hui Lin
- Department of Pathophysiology, School of Basic Medical Sciences, Nanchang University, Nanchang, 330006, Jiangxi, China.
| |
Collapse
|
290
|
González-Acedo A, Ramos-Torrecillas J, Illescas-Montes R, Costela-Ruiz VJ, Ruiz C, Melguizo-Rodríguez L, García-Martínez O. The Benefits of Olive Oil for Skin Health: Study on the Effect of Hydroxytyrosol, Tyrosol, and Oleocanthal on Human Fibroblasts. Nutrients 2023; 15:2077. [PMID: 37432217 DOI: 10.3390/nu15092077] [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: 03/21/2023] [Revised: 04/20/2023] [Accepted: 04/21/2023] [Indexed: 07/12/2023] Open
Abstract
Fibroblasts contribute to maintaining tissue integrity and homeostasis and are a key cell population in wound healing. This cell population can be stimulated by some bioactive compounds such as extra virgin olive oil (EVOO) polyphenols. The aim of this study was to determine the effects of hydroxytyrosol (htyr), tyrosol (tyr), and oleocanthal (ole) phenolic compounds present in EVOO on the proliferation, migration, cell cycle, and antigenic profile of cultured human fibroblasts. CCD-1064Sk human fibroblast cells were treated for 24 h with each polyphenol at doses ranging 10-5 to 10-9 M. Cell proliferation was evaluated using the MTT spectrophotometric technique, migration capacity by culture insert assay, and cell cycle and antigenic profile with flow cytometry. Cell proliferation was significantly increased by treatment with all compounds. The highest increases followed treatments with htyr or tyr at doses of 10-5 or 10-6 M and with ole at 10-6 and 10-7 M, and these compounds and doses were used for assays of antigenic profile, cell cycle, and migration. During the first few hours after treatment, increased fibronectin and α-actin expressions and greater cell migration were observed, with no cell cycle changes. In conclusion, these in vitro results suggest that phenolic compounds in EVOO might contribute to wound healing through action on fibroblasts related to tissue regeneration.
Collapse
Affiliation(s)
- Anabel González-Acedo
- Biomedical Group (BIO277), Department of Nursing, Faculty of Health Sciences, University of Granada, C/Santander, 1, 52005 Melilla, Spain
| | - Javier Ramos-Torrecillas
- Biomedical Group (BIO277), Department of Nursing, Faculty of Health Sciences, University of Granada, Avda. Ilustración 60, 18016 Granada, Spain
- Institute of Biosanitary Research, ibs.Granada, C/Doctor Azpitarte 4, 4a Planta, 18012 Granada, Spain
| | - Rebeca Illescas-Montes
- Biomedical Group (BIO277), Department of Nursing, Faculty of Health Sciences, University of Granada, Avda. Ilustración 60, 18016 Granada, Spain
- Institute of Biosanitary Research, ibs.Granada, C/Doctor Azpitarte 4, 4a Planta, 18012 Granada, Spain
| | - Víctor J Costela-Ruiz
- Institute of Biosanitary Research, ibs.Granada, C/Doctor Azpitarte 4, 4a Planta, 18012 Granada, Spain
- Biomedical Group (BIO277), Department of Nursing, Faculty of Health Sciences, University of Granada, C/Cortadura del Valle, s.n., 51001 Ceuta, Spain
| | - Concepción Ruiz
- Biomedical Group (BIO277), Department of Nursing, Faculty of Health Sciences, University of Granada, Avda. Ilustración 60, 18016 Granada, Spain
- Institute of Biosanitary Research, ibs.Granada, C/Doctor Azpitarte 4, 4a Planta, 18012 Granada, Spain
- Institute of Neuroscience, Centro de Investigación Biomédica (CIBM), University of Granada, Parque de Tecnológico de la Salud (PTS) Avda. del Conocimiento S/N, Armilla, 18016 Granada, Spain
| | - Lucía Melguizo-Rodríguez
- Biomedical Group (BIO277), Department of Nursing, Faculty of Health Sciences, University of Granada, Avda. Ilustración 60, 18016 Granada, Spain
- Institute of Biosanitary Research, ibs.Granada, C/Doctor Azpitarte 4, 4a Planta, 18012 Granada, Spain
| | - Olga García-Martínez
- Biomedical Group (BIO277), Department of Nursing, Faculty of Health Sciences, University of Granada, Avda. Ilustración 60, 18016 Granada, Spain
- Institute of Biosanitary Research, ibs.Granada, C/Doctor Azpitarte 4, 4a Planta, 18012 Granada, Spain
| |
Collapse
|
291
|
Costa BH, Rezende AK, Costa L, Neves GFM, Shimano AC, de Oliveira Penoni Á, Carvalho CR, Costa RA, de Alvarenga EC. Systemic effects of oral tolerance in bone healing. Sci Rep 2023; 13:6296. [PMID: 37072616 PMCID: PMC10113372 DOI: 10.1038/s41598-023-33591-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2022] [Accepted: 04/15/2023] [Indexed: 05/03/2023] Open
Abstract
Bone fractures cause acute inflammation that, despite being important for initial repair, may delay the healing of the damaged bone. Parenteral injection of dietary protein has been shown to decrease inflammation and accelerate the repair of skin wounds and other inflammatory pathologies. Thus, our aim was to evaluate whether the intraperitoneal (i.p.) immunization with zein, an abundant protein in rodent chow, would favor bone healing. Wistar rats received i.p. immunization: saline (SG), adjuvant (AG) and zein associated with adjuvant (ZG). Then, a 2 mm of defect bone was performed on the right tibia, and on days 7, 14, 28 and 45 thereafter, analyses were performed. The results showed that the injection of zein reduced inflammation without impairing bone mineralization. Moreover, biomechanical tests demonstrated higher levels of maximum force (N) in ZG, indicating better mechanical resistance in relation to the others. The computerized tomography also indicated lower levels of medullary content in the ZG than in the SG, suggesting the absence of trabeculae in the medullary region in the ZG. These findings suggest that the injection of zein in previously tolerated animals may improve bone repair, leading to mechanically functional bone formation.
Collapse
Affiliation(s)
- Bruno Henrique Costa
- Departamento de Ciências Naturais, Universidade Federal de São João Del Rei, Praça Dom Helvécio 74, São João del Rei, Minas Gerais, 36301-160, Brazil
| | - Alisson Kennedy Rezende
- Departamento de Ciências Naturais, Universidade Federal de São João Del Rei, Praça Dom Helvécio 74, São João del Rei, Minas Gerais, 36301-160, Brazil
| | - Lais Costa
- Departamento de Ciências Naturais, Universidade Federal de São João Del Rei, Praça Dom Helvécio 74, São João del Rei, Minas Gerais, 36301-160, Brazil
| | - Gabrielle Fernanda Monteiro Neves
- Departamento de Ciências Naturais, Universidade Federal de São João Del Rei, Praça Dom Helvécio 74, São João del Rei, Minas Gerais, 36301-160, Brazil
| | - Antônio Carlos Shimano
- Departamento de Biomecânica, Medicina e Reabilitação do Aparelho Locomotor, Faculdade de Medicina de Ribeirão Preto, Universidade de São Paulo, Ribeirão Preto, São Paulo, 14049-900, Brazil
| | - Álvaro de Oliveira Penoni
- Departamento das Ciências da Educação Física e Saúde, Universidade Federal de São João del-Rei, São João del Rei, Minas Gerais, 36301-360, Brazil
| | - Claudia Rocha Carvalho
- Departamento de Ciências Naturais, Universidade Federal de São João Del Rei, Praça Dom Helvécio 74, São João del Rei, Minas Gerais, 36301-160, Brazil
- Departamento Morfologia, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais, Belo Horizonte, Minas Gerais, 31270-901, Brazil
| | - Raquel Alves Costa
- Departamento de Ciências Naturais, Universidade Federal de São João Del Rei, Praça Dom Helvécio 74, São João del Rei, Minas Gerais, 36301-160, Brazil
| | - Erika Costa de Alvarenga
- Departamento de Ciências Naturais, Universidade Federal de São João Del Rei, Praça Dom Helvécio 74, São João del Rei, Minas Gerais, 36301-160, Brazil.
| |
Collapse
|
292
|
Fabre T, Barron AMS, Christensen SM, Asano S, Bound K, Lech MP, Wadsworth MH, Chen X, Wang C, Wang J, McMahon J, Schlerman F, White A, Kravarik KM, Fisher AJ, Borthwick LA, Hart KM, Henderson NC, Wynn TA, Dower K. Identification of a broadly fibrogenic macrophage subset induced by type 3 inflammation. Sci Immunol 2023; 8:eadd8945. [PMID: 37027478 DOI: 10.1126/sciimmunol.add8945] [Citation(s) in RCA: 125] [Impact Index Per Article: 62.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/09/2023]
Abstract
Macrophages are central orchestrators of the tissue response to injury, with distinct macrophage activation states playing key roles in fibrosis progression and resolution. Identifying key macrophage populations found in human fibrotic tissues could lead to new treatments for fibrosis. Here, we used human liver and lung single-cell RNA sequencing datasets to identify a subset of CD9+TREM2+ macrophages that express SPP1, GPNMB, FABP5, and CD63. In both human and murine hepatic and pulmonary fibrosis, these macrophages were enriched at the outside edges of scarring and adjacent to activated mesenchymal cells. Neutrophils expressing MMP9, which participates in the activation of TGF-β1, and the type 3 cytokines GM-CSF and IL-17A coclustered with these macrophages. In vitro, GM-CSF, IL-17A, and TGF-β1 drive the differentiation of human monocytes into macrophages expressing scar-associated markers. Such differentiated cells could degrade collagen IV but not collagen I and promote TGF-β1-induced collagen I deposition by activated mesenchymal cells. In murine models blocking GM-CSF, IL-17A or TGF-β1 reduced scar-associated macrophage expansion and hepatic or pulmonary fibrosis. Our work identifies a highly specific macrophage population to which we assign a profibrotic role across species and tissues. It further provides a strategy for unbiased discovery, triage, and preclinical validation of therapeutic targets based on this fibrogenic macrophage population.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | | | | | - Ju Wang
- Pfizer Inc., Cambridge, MA, USA
| | | | | | | | | | - Andrew J Fisher
- Translational and Clinical Research Institute, Faculty of Medical Sciences, Newcastle University, Newcastle upon Tyne, UK
| | - Lee A Borthwick
- Newcastle Fibrosis Research Group, Biosciences Institute, Newcastle University, Newcastle upon Tyne, UK
| | | | - Neil C Henderson
- MRC Human Genetics Unit, Institute of Genetics and Cancer, University of Edinburgh, Edinburgh, UK
- Centre for Inflammation Research, the Queen's Medical Research Institute, University of Edinburgh, Edinburgh, UK
| | | | | |
Collapse
|
293
|
Xu M, Ji X, Huo J, Chen J, Liu N, Li Z, Jia Q, Sun B, Zhu M, Li P. Nonreleasing AgNP Colloids Composite Hydrogel with Potent Hemostatic, Photodynamic Bactericidal and Wound Healing-Promoting Properties. ACS APPLIED MATERIALS & INTERFACES 2023; 15:17742-17756. [PMID: 37006134 DOI: 10.1021/acsami.3c03247] [Citation(s) in RCA: 14] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/19/2023]
Abstract
Reactive oxygen species (ROS) produced by noble metallic nanoparticles under visible light is an effective way to combat drug-resistant bacteria colonized on the wound. However, the photocatalytic efficiency of noble metallic nanoparticles is limited by its self-aggregation in water media. Moreover, the fast release of noble metallic ions from nanoparticles might engender cellular toxicity and hazardous environmental issues. Herein, we chose AgNPs, the most common plasmonic noble metallic nanoparticles, as an example, modifying the surface of AgNPs with oleic acid and n-butylamine and imbedded them into calcium alginate (CA) hydrogel that holds tissue adhesion, rapid hemostatic, sunlight-sensitive antibacterial and anti-inflammatory abilities, and thus effectively promotes the healing of wounds. Unlike conventional AgNP-based materials, the constrain of colloids and hydrogel networks hinders the leach of Ag+. Nonetheless, the CA/Ag hydrogels exhibit on-demand photodynamic antibacterial efficacy due to the generation of ROS under visible light. In addition, the CA/Ag hydrogel can effectively stop the hemorrhage in a mouse liver bleeding model due to their skin-adaptive flexibility and tissue adhesiveness. The potent sunlight-responsive antibacterial activity of the CA/Ag hydrogel can effectively kill multidrug-resistant bacteria both in vitro (>99.999%) and in vivo (>99.9%), while the diminished Ag+ release guarantees its biocompatibility. The CA/Ag hydrogel significantly promotes the wound healing process by the downregulation of proinflammatory cytokines (TNF-α and IL-6) in a rodent full-thickness cutaneous wound model. Overall, the proposed multifunctional CA/Ag nanocomposite hydrogel has excellent prospects as an advanced wound dressing.
Collapse
Affiliation(s)
- Miao Xu
- Frontiers Science Center for Flexible Electronics (FSCFE), Xi'an Institute of Flexible Electronics (IFE) & Xi'an Institute of Biomedical Materials and Engineering (IBME), Northwestern Polytechnical University (NPU), Xi'an 710072, China
- Key Laboratory of Flexible Electronics (KLOFE) and Institute of Advanced Materials (IAM), Jiangsu National Synergetic Innovation Center for Advanced Materials (SICAM), Nanjing Tech University, Nanjing 211816, China
| | - Xiaohuan Ji
- State Key Laboratory for Modification of Chemical Fibers and Polymer Materials, College of Materials Science and Engineering, Donghua University, Shanghai 201620, P. R. China
| | - Jingjing Huo
- Frontiers Science Center for Flexible Electronics (FSCFE), Xi'an Institute of Flexible Electronics (IFE) & Xi'an Institute of Biomedical Materials and Engineering (IBME), Northwestern Polytechnical University (NPU), Xi'an 710072, China
| | - Jingjie Chen
- Frontiers Science Center for Flexible Electronics (FSCFE), Xi'an Institute of Flexible Electronics (IFE) & Xi'an Institute of Biomedical Materials and Engineering (IBME), Northwestern Polytechnical University (NPU), Xi'an 710072, China
| | - Nian Liu
- Frontiers Science Center for Flexible Electronics (FSCFE), Xi'an Institute of Flexible Electronics (IFE) & Xi'an Institute of Biomedical Materials and Engineering (IBME), Northwestern Polytechnical University (NPU), Xi'an 710072, China
| | - Ziyue Li
- Key Laboratory of Flexible Electronics (KLOFE) and Institute of Advanced Materials (IAM), Jiangsu National Synergetic Innovation Center for Advanced Materials (SICAM), Nanjing Tech University, Nanjing 211816, China
| | - Qingyan Jia
- Frontiers Science Center for Flexible Electronics (FSCFE), Xi'an Institute of Flexible Electronics (IFE) & Xi'an Institute of Biomedical Materials and Engineering (IBME), Northwestern Polytechnical University (NPU), Xi'an 710072, China
| | - Bin Sun
- State Key Laboratory for Modification of Chemical Fibers and Polymer Materials, College of Materials Science and Engineering, Donghua University, Shanghai 201620, P. R. China
| | - Meifang Zhu
- State Key Laboratory for Modification of Chemical Fibers and Polymer Materials, College of Materials Science and Engineering, Donghua University, Shanghai 201620, P. R. China
| | - Peng Li
- Frontiers Science Center for Flexible Electronics (FSCFE), Xi'an Institute of Flexible Electronics (IFE) & Xi'an Institute of Biomedical Materials and Engineering (IBME), Northwestern Polytechnical University (NPU), Xi'an 710072, China
- Key Laboratory of Flexible Electronics (KLOFE) and Institute of Advanced Materials (IAM), Jiangsu National Synergetic Innovation Center for Advanced Materials (SICAM), Nanjing Tech University, Nanjing 211816, China
- The State Key Laboratory of Molecular Engineering of Polymers, Department of Macromolecular Science, Fudan University, Shanghai 200438, China
| |
Collapse
|
294
|
Wang X, Zhu X, Wang D, Li X, Wang J, Yin G, Huang Z, Pu X. Identification of a Specific Phage as Growth Factor Alternative Promoting the Recruitment and Differentiation of MSCs in Bone Tissue Regeneration. ACS Biomater Sci Eng 2023; 9:2426-2437. [PMID: 37023478 DOI: 10.1021/acsbiomaterials.2c01538] [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] [Indexed: 04/08/2023]
Abstract
Inefficient use and loss of exogenously implanted mesenchymal stem cells (MSCs) are major concerns in MSCs-based bone tissue engineering. It is a promising approach to overcome the above issues by recruiting and regulation of endogenous MSCs. However, there are few substances that can recruit MSCs effectively and specifically to the site of bone injury. In this study, we identified a phage clone (termed P11) with specific affinity for MSCs through phage display biopanning, and further investigated the effects of P11 on the cytological behavior of MSCs and macrophages. The results showed that P11 could bind MSCs specifically and promote the proliferation and migration of MSCs. Meanwhile, P11 could polarize macrophages to the M1 phenotype and significantly changed their morphology, which further enhanced the chemotaxis of MSCs. Additionally, RNA-seq results revealed that P11 could promote the secretion of osteogenesis-related markers in MSCs through the TPL2-MEK-ERK signaling pathway. Altogether, P11 has great potential to be used as growth factor alternatives in bone tissue engineering, with the advantages of cheaper and stable activity. Our study also advances the understanding of the effects of phages on macrophages and MSCs, and provides a new idea for the development in the field of phage-based tissue engineering.
Collapse
Affiliation(s)
- Xingming Wang
- College of Biomedical Engineering, Sichuan University, Chengdu 610065, People's Republic of China
| | - Xiupeng Zhu
- Institute of Biomedical Engineering, College of Medicine, Southwest Jiaotong University, Chengdu 610031, People's Republic of China
- Key Laboratory of Advanced Technologies of Materials Ministry of Education, School of Materials Science and Engineering, Southwest Jiaotong University, Chengdu 610031, People's Republic of China
| | - Danni Wang
- College of Biomedical Engineering, Sichuan University, Chengdu 610065, People's Republic of China
| | - Xiaoxu Li
- College of Biomedical Engineering, Sichuan University, Chengdu 610065, People's Republic of China
| | - Juan Wang
- College of Biomedical Engineering, Sichuan University, Chengdu 610065, People's Republic of China
| | - Guangfu Yin
- College of Biomedical Engineering, Sichuan University, Chengdu 610065, People's Republic of China
| | - Zhongbing Huang
- College of Biomedical Engineering, Sichuan University, Chengdu 610065, People's Republic of China
| | - Ximing Pu
- College of Biomedical Engineering, Sichuan University, Chengdu 610065, People's Republic of China
| |
Collapse
|
295
|
Bencivenga D, Arcadio F, Piccirillo A, Annunziata M, Della Ragione F, Cennamo N, Borriello A, Zeni L, Guida L. Plasmonic optical fiber biosensor development for point-of-care detection of malondialdehyde as a biomarker of oxidative stress. Free Radic Biol Med 2023; 199:177-188. [PMID: 36841362 DOI: 10.1016/j.freeradbiomed.2023.02.020] [Citation(s) in RCA: 20] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/12/2022] [Revised: 02/07/2023] [Accepted: 02/21/2023] [Indexed: 02/26/2023]
Abstract
Numerous pieces of evidence demonstrate that oxidative stress impairs biological functions, speeds up aging, and has a role in a variety of human diseases, including systemic and oral inflammatory disorders, and even cancer. Therefore, technologies providing accurate measures of oxidative stress indicators or biomarkers appear essential in the identification/prevention of such diseases, and in their management. Particularly advantageous is the employement of point-of-care tests based on affordable and small biochips since they can quickly process biological samples and deliver results near the point of care for a prompt therapeutic intervention. Malondialdehyde (MDA) is a key byproduct of oxidative reaction and has been identified as an effective marker of oxidative stress. Herein, we describe the detection of MDA in buffer and in a complex matrix such as saliva, using a plasmonic optical fiber device combined with a highly selective anti-MDA antibody. The experimental results highlight the excellent performance of the proposed biosensor, as well as its ability to provide a low-cost point-of-care test (PoC-T) to be used in real life situations. We demonstrated that a single saliva dilution step and a short incubation time are required for the accurate detection of low concentrations of total MDA (free and conjugated). As a proof-of-concept of future biomedical applications, the method has been tested to determine MDA concentration in saliva of a periodontitis patient compared to that of a healthy control. The obtained findings represent the basis for developing PoC-Ts to be employed in monitoring oral diseases like periodontitis, oral cancers or systemic oxidative-stress associated pathologies. Conclusively, our study puts the ground for an oxidative stress biosensor widely-applicable to different scenarios.
Collapse
Affiliation(s)
- Debora Bencivenga
- Department of Precision Medicine, University of Campania "Luigi Vanvitelli", Via de Crecchio, 7 - 80138, Naples, Italy
| | - Francesco Arcadio
- Department of Engineering, University of Campania "Luigi Vanvitelli", Via Roma, 9 - 81031, Aversa, (CE), Italy
| | - Angelantonio Piccirillo
- Multidisciplinary Department of Medical-Surgical and Dental Specialties, University of Campania "Luigi Vanvitelli", via De Crecchio, 6 - 80138, Naples, Italy
| | - Marco Annunziata
- Multidisciplinary Department of Medical-Surgical and Dental Specialties, University of Campania "Luigi Vanvitelli", via De Crecchio, 6 - 80138, Naples, Italy.
| | - Fulvio Della Ragione
- Department of Precision Medicine, University of Campania "Luigi Vanvitelli", Via de Crecchio, 7 - 80138, Naples, Italy
| | - Nunzio Cennamo
- Department of Engineering, University of Campania "Luigi Vanvitelli", Via Roma, 9 - 81031, Aversa, (CE), Italy.
| | - Adriana Borriello
- Department of Precision Medicine, University of Campania "Luigi Vanvitelli", Via de Crecchio, 7 - 80138, Naples, Italy.
| | - Luigi Zeni
- Department of Engineering, University of Campania "Luigi Vanvitelli", Via Roma, 9 - 81031, Aversa, (CE), Italy
| | - Luigi Guida
- Multidisciplinary Department of Medical-Surgical and Dental Specialties, University of Campania "Luigi Vanvitelli", via De Crecchio, 6 - 80138, Naples, Italy
| |
Collapse
|
296
|
Wu L, Huang K, Li Q, Wu H, Gao Y, Xu X, Liu X, Han L. Crosstalk between myofibroblasts and macrophages: A regulative factor of valvular fibrosis in calcific aortic valve disease. Cell Biol Int 2023; 47:754-767. [PMID: 36542640 DOI: 10.1002/cbin.11980] [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: 09/02/2022] [Revised: 11/16/2022] [Accepted: 12/08/2022] [Indexed: 12/24/2022]
Abstract
Inflammation and fibrosis are highly correlated with the progression of calcific aortic valve disease (CAVD). As one of the differentiated forms of valvular interstitial cells, myofibroblasts play a critical role in CAVD's development as do macrophages. Although numerous studies have been conducted on them separately, their communication and interaction remain unclear. We used porcine aortic valves to isolate valve interstitial cells (VICs). VICs were induced to differentiate into myofibroblasts by transforming growth factor-β1 (TGF-β1). After successful activation was determined, the myofibroblast-conditioned medium (CM) was collected and used to act on RAW264.7, a macrophage cell line. A migration and adhesion assay estimated the recruitment capability of myofibroblasts on macrophages. We used flow cytometry, quantitative polymerase chain reaction (qPCR), and Western blot analysis to investigate myofibroblasts' polarity promotion function in macrophages. Finally, we used macrophage-CM on VICs to explore the differentiation induction function of polarized macrophages. Myofibroblast marker alpha-smooth muscle actin and M2 macrophage marker CD163 were detected as upregulated in CAVD patients, and their expression has a certain correlation. The Smad3/HA/CD44 axis activated the differentiation of myofibroblasts by Western blot. The myofibroblast-CM can promote chemotaxis and adhesion of macrophages through protein kinase B/chemokine (C-C motif) ligand5 and Smad3/HA/CD44, respectively. Hyaluronic acid (HA) inside the myofibroblast-CM stimulates macrophages to polarize into M2 macrophages. In turn, M2 macrophage-CM has the promotive ability to activate myofibroblasts but fails to induce the osteoblast differentiation of VICs directly. The crosstalk between myofibroblasts and macrophages causes the excessive activation of myofibroblasts. This positive feedback loop may play a vital role in CAVD progression.
Collapse
Affiliation(s)
- Lujia Wu
- Department of Cardiothoracic Surgery, Changhai Hospital, Second Military Medical University, Shanghai, China
| | - Kai Huang
- Department of Cardiothoracic Surgery, Changhai Hospital, Second Military Medical University, Shanghai, China
| | - Qin Li
- Department of Cardiothoracic Surgery, Changhai Hospital, Second Military Medical University, Shanghai, China
| | - Hao Wu
- Department of Cardiothoracic Surgery, Changhai Hospital, Second Military Medical University, Shanghai, China
| | - Yuan Gao
- Department of Cardiothoracic Surgery, Changhai Hospital, Second Military Medical University, Shanghai, China
| | - Xiangyang Xu
- Department of Cardiothoracic Surgery, Changhai Hospital, Second Military Medical University, Shanghai, China
| | - Xiaohong Liu
- Department of Cardiothoracic Surgery, Changhai Hospital, Second Military Medical University, Shanghai, China
| | - Lin Han
- Department of Cardiothoracic Surgery, Changhai Hospital, Second Military Medical University, Shanghai, China
| |
Collapse
|
297
|
Long L, Ji D, Hu C, Yang L, Tang S, Wang Y. Microneedles for in situ tissue regeneration. Mater Today Bio 2023; 19:100579. [PMID: 36880084 PMCID: PMC9984687 DOI: 10.1016/j.mtbio.2023.100579] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2022] [Revised: 02/06/2023] [Accepted: 02/10/2023] [Indexed: 02/13/2023] Open
Abstract
Tissue injury is a common clinical problem, which may cause great burden on patients' life. It is important to develop functional scaffolds to promote tissue repair and regeneration. Due to their unique composition and structure, microneedles have attracted extensive attention in various tissues regeneration, including skin wound, corneal injury, myocardial infarction, endometrial injury, and spinal cord injury et al. Microneedles with micro-needle structure can effectively penetrate the barriers of necrotic tissue or biofilm, therefore improving the bioavailability of drugs. The use of microneedles to deliver bioactive molecules, mesenchymal stem cells, and growth factors in situ allows for targeted tissue and better spatial distribution. At the same time, microneedles can also provide mechanical support or directional traction for tissue, thus accelerating tissue repair. This review summarized the research progress of microneedles for in situ tissue regeneration over the past decade. At the same time, the shortcomings of existing researches, future research direction and clinical application prospect were also discussed.
Collapse
Affiliation(s)
- Linyu Long
- Aier Eye Institute, Changsha, Hunan Province, 410035, China
- Eye Center of Xiangya Hospital, Central South University, Changsha, 410008, Hunan, China
| | - Dan Ji
- Eye Center of Xiangya Hospital, Central South University, Changsha, 410008, Hunan, China
| | - Cheng Hu
- National Engineering Research Center for Biomaterials, Sichuan University, Chengdu, 610064, China
| | - Li Yang
- National Engineering Research Center for Biomaterials, Sichuan University, Chengdu, 610064, China
| | - Shibo Tang
- Aier Eye Institute, Changsha, Hunan Province, 410035, China
- Aier School of Ophthalmology, Central South University, Changsha, Hunan, 410009, China
- CAS Center for Excellence in Brain Science and Intelligence Technology, Chinese Academy of Sciences, Shanghai, 200031, China
| | - Yunbing Wang
- National Engineering Research Center for Biomaterials, Sichuan University, Chengdu, 610064, China
| |
Collapse
|
298
|
Zhang Y, Wang L, Kang H, Lin CY, Fan Y. Unlocking the Therapeutic Potential of Irisin: Harnessing Its Function in Degenerative Disorders and Tissue Regeneration. Int J Mol Sci 2023; 24:ijms24076551. [PMID: 37047523 PMCID: PMC10095399 DOI: 10.3390/ijms24076551] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2023] [Revised: 03/26/2023] [Accepted: 03/28/2023] [Indexed: 04/03/2023] Open
Abstract
Physical activity is well-established as an important protective factor against degenerative conditions and a promoter of tissue growth and renewal. The discovery of Fibronectin domain-containing protein 5 (FNDC5) as the precursor of Irisin in 2012 sparked significant interest in its potential as a diagnostic biomarker and a therapeutic agent for various diseases. Clinical studies have examined the correlation between plasma Irisin levels and pathological conditions using a range of assays, but the lack of reliable measurements for endogenous Irisin has led to uncertainty about its prognostic/diagnostic potential as an exercise surrogate. Animal and tissue-engineering models have shown the protective effects of Irisin treatment in reversing functional impairment and potentially permanent damage, but dosage ambiguities remain unresolved. This review provides a comprehensive examination of the clinical and basic studies of Irisin in the context of degenerative conditions and explores its potential as a therapeutic approach in the physiological processes involved in tissue repair/regeneration.
Collapse
Affiliation(s)
- Yuwei Zhang
- Key Laboratory for Biomechanics and Mechanobiology of Ministry of Education, Beijing Advanced Innovation Centre for Biomedical Engineering, School of Biological Science and Medical Engineering, Beihang University, Beijing 100083, China
| | - Lizhen Wang
- Key Laboratory for Biomechanics and Mechanobiology of Ministry of Education, Beijing Advanced Innovation Centre for Biomedical Engineering, School of Biological Science and Medical Engineering, Beihang University, Beijing 100083, China
- Correspondence:
| | - Hongyan Kang
- Key Laboratory for Biomechanics and Mechanobiology of Ministry of Education, Beijing Advanced Innovation Centre for Biomedical Engineering, School of Biological Science and Medical Engineering, Beihang University, Beijing 100083, China
| | - Chia-Ying Lin
- Key Laboratory for Biomechanics and Mechanobiology of Ministry of Education, Beijing Advanced Innovation Centre for Biomedical Engineering, School of Biological Science and Medical Engineering, Beihang University, Beijing 100083, China
- Department of Biomedical, Chemical & Environmental Engineering, University of Cincinnati, Cincinnati, OH 45267, USA
- Department of Orthopaedic Surgery, University of Cincinnati, Cincinnati, OH 45267, USA
- Department of Neurosurgery, University of Cincinnati, Cincinnati, OH 45267, USA
| | - Yubo Fan
- Key Laboratory for Biomechanics and Mechanobiology of Ministry of Education, Beijing Advanced Innovation Centre for Biomedical Engineering, School of Biological Science and Medical Engineering, Beihang University, Beijing 100083, China
- School of Engineering Medicine, Beihang University, Beijing 100083, China
| |
Collapse
|
299
|
Michels M, Córneo E, Rocha LBG, Dias R, Voytena APL, Rossetto M, Ramlov F, Dal-Pizzol F, Jesus GFA. Paraprobiotics strains accelerate wound repair by stimulating re-epithelialization of NIH-3T3 cells, decreasing inflammatory response and oxidative stress. Arch Microbiol 2023; 205:134. [PMID: 36959516 DOI: 10.1007/s00203-023-03469-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2022] [Accepted: 03/06/2023] [Indexed: 03/25/2023]
Abstract
The present study aimed to evaluate the potential and specificity of the inflammatory and antioxidant response of Microbe-Associated Molecular Patterns (MAMPs) in NIH-3T3 fibroblast cells, as well as in the healing process of skin wounds. Cells (NIH-3T3) were cultivated in supplemented specific medium. NIH-3T3 cells were treated with MAMPs (Bifidobacterium lactis or Lactobacillus casei or Lactobacillus gasseri or Lactobacillus paracasei or Streptococcus thermophilus), at two concentrations and insulted with LPS or H2O2. Cell viability, myeloperoxidase activity, nitrite/nitrate, oxidative damage and inflammatory parameters were measured. In addition, scratch assay was performed. Significant scratch closure was observed after 24 h and 48 h, and the effect of 0.1 g/mL MAMPs on wound healing was found to be highly statistically significant. In the viability cellular assay, Lactobacillus showed better response in 0.1 g/mL dose, whereas B. lactis and S. thermophilus showed better response in 0.01 g/mL dose. There was reduction in IL-6 and IL-1β levels in all treatments insulted with LPS. MAMP's showed preventive efficacy in reducing the effects caused by LPS. The MAMP's action in decreasing the production of ROS, inflammatory activity and increasing cell viability, besides significant cell proliferation during wound healing processes suggests remodeling mechanisms and new possibilities for wound healing.
Collapse
Affiliation(s)
- Monique Michels
- Gabbia Biotechnology, Barra Velha, Santa Catarina, Brazil.
- Biohall Research and Innovation, Itajaí, Santa Catarina, Brazil.
| | - Emily Córneo
- Laboratory of Experimental Pathophysiology-Graduate Program in Health Sciences, University of Southern Santa Catarina (UNESC), Criciúma, Brazil
| | - Luana Bezerra Gonçalves Rocha
- Laboratory of Experimental Pathophysiology-Graduate Program in Health Sciences, University of Southern Santa Catarina (UNESC), Criciúma, Brazil
| | - Rodrigo Dias
- Laboratory of Experimental Pathophysiology-Graduate Program in Health Sciences, University of Southern Santa Catarina (UNESC), Criciúma, Brazil
| | | | | | | | - Felipe Dal-Pizzol
- Laboratory of Experimental Pathophysiology-Graduate Program in Health Sciences, University of Southern Santa Catarina (UNESC), Criciúma, Brazil
| | - Gabriel Fernandes Alves Jesus
- Gabbia Biotechnology, Barra Velha, Santa Catarina, Brazil
- Biohall Research and Innovation, Itajaí, Santa Catarina, Brazil
| |
Collapse
|
300
|
Anti-Inflammatory Effect and Toxicological Profile of Pulp Residue from the Caryocar Brasiliense, a Sustainable Raw Material. Gels 2023; 9:gels9030234. [PMID: 36975683 PMCID: PMC10048353 DOI: 10.3390/gels9030234] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2022] [Revised: 12/28/2022] [Accepted: 02/02/2023] [Indexed: 03/19/2023] Open
Abstract
Caryocar brasiliense Cambess is a plant species typical of the Cerrado, a Brazilian biome. The fruit of this species is popularly known as pequi, and its oil is used in traditional medicine. However, an important factor hindering the use of pequi oil is its low yield when extracted from the pulp of this fruit. Therefore, in this study, with aim of developing a new herbal medicine, we an-alyzed the toxicity and anti-inflammatory activity of an extract of pequi pulp residue (EPPR), fol-lowing the mechanical extraction of the oil from its pulp. For this purpose, EPPR was prepared and encapsulated in chitosan. The nanoparticles were analyzed, and the cytotoxicity of the encapsu-lated EPPR was evaluated in vitro. After confirming the cytotoxicity of the encapsulated EPPR, the following evaluations were performed with non-encapsulated EPPR: in vitro anti-inflammatory activity, quantification of cytokines, and acute toxicity in vivo. Once the anti-inflammatory activity and absence of toxicity of EPPR were verified, a gel formulation of EPPR was developed for topical use and analyzed for its in vivo anti-inflammatory potential, ocular toxicity, and previous stability assessment. EPPR and the gel containing EPPR showed effective anti-inflammatory activity and lack of toxicity. The formulation was stable. Thus, a new herbal medicine with anti-inflammatory activity can be developed from discarded pequi residue.
Collapse
|