201
|
Lohuis PJFM, Maldonado-Chapa F, Santos-Santillana KM, Filipović B, Dirven R, Karakullukcku MB, Karssemakers L, Schreuder WH, Zuur CL, Timmermans J. Optimizing Wound Care after Surgery of the Head and Neck: A Review of Dressing Materials. Facial Plast Surg 2024; 40:68-79. [PMID: 36878677 DOI: 10.1055/a-2047-6356] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/08/2023] Open
Abstract
Wound healing is a complex biological process subject to complications that might jeopardize the patient's postoperative care. Appropriately approaching surgical wounds after head and neck surgery positively influences the quality and speed of wound healing and increases patient comfort. A large variety of dressing materials currently exist that allow the care of different types of wounds. Nevertheless, there is limited literature on the most suitable types of dressings after head and neck surgery. The objective of the present article is to review the most commonly used wound dressings, their benefits, indications, and disadvantages, and to provide a systematic approach for wound care within the head and neck. The Woundcare Consultant Society distinguishes wounds into three groups: black, yellow, and red. Each type of wound represents distinctive underlying pathophysiological processes with unique needs. Utilizing this classification along with the TIME model allows a proper characterization of wounds and the identification of potential healing barriers. This evidence-based and systematic approach can facilitate and guide the head and neck surgeon in selecting a wound dressing upon acknowledging their properties, which are herein reviewed and exemplified with representative cases.
Collapse
Affiliation(s)
- Peter J F M Lohuis
- Department of Head and Neck Surgery and Oncology, The Netherlands Cancer Institute, Amsterdam, The Netherlands
- Department of Facial Plastic and Reconstructive Surgery, Lohuis-Filipović Medial Group, Zagreb, Croatia
- Department of Facial Plastic and Reconstructive Surgery, Bergman Clinics, Hilversum, The Netherlands
- Department of Sleep Medicine, Ruysdael Clinics, Amsterdam, The Netherlands
| | - Felix Maldonado-Chapa
- Department of Head and Neck Surgery and Oncology, The Netherlands Cancer Institute, Amsterdam, The Netherlands
- Department of Facial Plastic and Reconstructive Surgery, Lohuis-Filipović Medial Group, Zagreb, Croatia
| | - Karla M Santos-Santillana
- Department of Facial Plastic and Reconstructive Surgery, Lohuis-Filipović Medial Group, Zagreb, Croatia
| | - Boris Filipović
- Department of Head and Neck Surgery and Oncology, The Netherlands Cancer Institute, Amsterdam, The Netherlands
- Department of Facial Plastic and Reconstructive Surgery, Lohuis-Filipović Medial Group, Zagreb, Croatia
| | - Richard Dirven
- Department of Head and Neck Surgery and Oncology, The Netherlands Cancer Institute, Amsterdam, The Netherlands
| | - M Baris Karakullukcku
- Department of Head and Neck Surgery and Oncology, The Netherlands Cancer Institute, Amsterdam, The Netherlands
| | - Luc Karssemakers
- Department of Head and Neck Surgery and Oncology, The Netherlands Cancer Institute, Amsterdam, The Netherlands
| | - Willem H Schreuder
- Department of Head and Neck Surgery and Oncology, The Netherlands Cancer Institute, Amsterdam, The Netherlands
| | - Charlotte L Zuur
- Department of Head and Neck Surgery and Oncology, The Netherlands Cancer Institute, Amsterdam, The Netherlands
| | - Jacqueline Timmermans
- Department of Head and Neck Surgery and Oncology, The Netherlands Cancer Institute, Amsterdam, The Netherlands
| |
Collapse
|
202
|
Chen D, Yang H, Yang L, Tang Y, Zeng H, He J, Chen W, Qu Y, Hu Y, Xu Y, Liu D, Song H, Li Q. Preoperative psychological symptoms and chronic postsurgical pain: analysis of the prospective China Surgery and Anaesthesia Cohort study. Br J Anaesth 2024; 132:359-371. [PMID: 37953200 DOI: 10.1016/j.bja.2023.10.015] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2023] [Revised: 10/10/2023] [Accepted: 10/11/2023] [Indexed: 11/14/2023] Open
Abstract
BACKGROUND Both preoperative psychological symptoms and chronic postsurgical pain (CPSP) are prevalent conditions and major concerns among surgery patients, with inconclusive associations. METHODS Based on the China Surgery and Anaesthesia Cohort (CSAC), we recruited 8350 surgery patients (40-65 yr old) from two medical centres between July 2020 and March 2023. Patients with preoperative psychological symptoms (i.e. anxiety, depression, stress reaction, and poor sleep quality) were identified using corresponding well-established scales. We then examined the associations of individual preoperative psychological symptoms and major patterns of preoperative psychological symptoms (identified by k-means clustering analysis) with CPSP, and different pain trajectories within 3 months. Lastly, mediation analyses were conducted to elucidate the mediating role of surgery/anaesthesia-related factors and the presence of 1-month postoperative psychological symptoms on the studied associations. RESULTS We included 1302 (1302/8350, 15.6%) CPSP patients. When analysed separately, all studied preoperative psychological symptoms were associated with increased CPSP risk, with the most pronounced odds ratio noted for anxiety (1.52, 95% confidence interval [CI] 1.23-1.86). Compared with patients clustered in the minor symptom group, excess risk of CPSP and experiencing an increasing pain trajectory was increased among patients with preoperative psychological symptoms featured by sleep disturbances (odds ratio=1.46, 95% CI 1.25-1.70 for CPSP and 1.58, 95% CI 1.20-2.08 for increasing pain trajectory) and multiple psychological symptoms (1.84 [95% CI 1.48-2.28] and 4.34 [95% CI 3.20-5.88]). Mediation analyses revealed acute/subacute postsurgical pain and psychological symptoms existing 1 month after surgery as notable mediators of the observed associations. CONCLUSIONS The presence of preoperative psychological symptoms might individually or jointly increase the risk of chronic postsurgical pain or experiencing deterioration in pain trajectory. Interventions for managing acute/subacute postsurgical pain and psychological symptoms at 1 month after surgery might help reduce such risk. CLINICAL TRIAL REGISTRATION ChiCTR2000034039.
Collapse
Affiliation(s)
- Dongxu Chen
- Department of Anaesthesiology and West China Biomedical Big Data Centre, West China Hospital, Sichuan University, Chengdu, China; Laboratory of Anaesthesia and Critical Care Medicine, National-Local Joint Engineering Research Centre of Translational Medicine of Anaesthesiology, West China Hospital, Sichuan University, Chengdu, China; West China Biomedical Big Data Centre, West China Hospital, Sichuan University, Chengdu, China
| | - Huazhen Yang
- West China Biomedical Big Data Centre, West China Hospital, Sichuan University, Chengdu, China; Med-X Center for Informatics, Sichuan University, Chengdu, China
| | - Lei Yang
- Department of Anaesthesiology and West China Biomedical Big Data Centre, West China Hospital, Sichuan University, Chengdu, China; Laboratory of Anaesthesia and Critical Care Medicine, National-Local Joint Engineering Research Centre of Translational Medicine of Anaesthesiology, West China Hospital, Sichuan University, Chengdu, China
| | - Yuling Tang
- Department of Anaesthesiology and West China Biomedical Big Data Centre, West China Hospital, Sichuan University, Chengdu, China; Laboratory of Anaesthesia and Critical Care Medicine, National-Local Joint Engineering Research Centre of Translational Medicine of Anaesthesiology, West China Hospital, Sichuan University, Chengdu, China
| | - Huolin Zeng
- Department of Anaesthesiology and West China Biomedical Big Data Centre, West China Hospital, Sichuan University, Chengdu, China; Laboratory of Anaesthesia and Critical Care Medicine, National-Local Joint Engineering Research Centre of Translational Medicine of Anaesthesiology, West China Hospital, Sichuan University, Chengdu, China
| | - Junhui He
- Department of Anaesthesiology and West China Biomedical Big Data Centre, West China Hospital, Sichuan University, Chengdu, China; Laboratory of Anaesthesia and Critical Care Medicine, National-Local Joint Engineering Research Centre of Translational Medicine of Anaesthesiology, West China Hospital, Sichuan University, Chengdu, China
| | - Wenwen Chen
- West China Biomedical Big Data Centre, West China Hospital, Sichuan University, Chengdu, China; Med-X Center for Informatics, Sichuan University, Chengdu, China
| | - Yuanyuan Qu
- West China Biomedical Big Data Centre, West China Hospital, Sichuan University, Chengdu, China; Med-X Center for Informatics, Sichuan University, Chengdu, China
| | - Yao Hu
- West China Biomedical Big Data Centre, West China Hospital, Sichuan University, Chengdu, China; Med-X Center for Informatics, Sichuan University, Chengdu, China
| | - Yueyao Xu
- West China Biomedical Big Data Centre, West China Hospital, Sichuan University, Chengdu, China; Med-X Center for Informatics, Sichuan University, Chengdu, China
| | - Di Liu
- West China Biomedical Big Data Centre, West China Hospital, Sichuan University, Chengdu, China; Med-X Center for Informatics, Sichuan University, Chengdu, China; Sichuan University - Pittsburgh Institute, Sichuan University, Chengdu, China
| | - Huan Song
- West China Biomedical Big Data Centre, West China Hospital, Sichuan University, Chengdu, China; Med-X Center for Informatics, Sichuan University, Chengdu, China; Centre of Public Health Sciences, Faculty of Medicine, University of Iceland, Reykjavík, Iceland.
| | - Qian Li
- Department of Anaesthesiology and West China Biomedical Big Data Centre, West China Hospital, Sichuan University, Chengdu, China; Laboratory of Anaesthesia and Critical Care Medicine, National-Local Joint Engineering Research Centre of Translational Medicine of Anaesthesiology, West China Hospital, Sichuan University, Chengdu, China.
| |
Collapse
|
203
|
Li B, Yang W, Shu R, Yang H, Yang F, Dai W, Chen W, Chan YK, Bai D, Deng Y. Antibacterial and Angiogenic (2A) Bio-Heterojunctions Facilitate Infectious Ischemic Wound Regeneration via an Endogenous-Exogenous Bistimulatory Strategy. ADVANCED MATERIALS (DEERFIELD BEACH, FLA.) 2024; 36:e2307613. [PMID: 37848208 DOI: 10.1002/adma.202307613] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/30/2023] [Revised: 09/23/2023] [Indexed: 10/19/2023]
Abstract
In infectious ischemic wounds, a lack of blood perfusion significantly worsens microbe-associated infection symptoms and frequently complicates healing. To overcome this daunting issue, antibacterial and angiogenic (2A) bio-heterojunctions (bio-HJs) consisting of CuS/MXene heterojunctions and a vascular endothelial growth factor (VEGF)-mimicking peptide (VMP) are devised and developed to accelerate infectious cutaneous regeneration by boosting angiogenesis via an endogenous-exogenous bistimulatory (EEB) strategy. Assisted by near-infrared irradiation, the bio-HJ platform exhibits versatile synergistic photothermal, photodynamic, and chemodynamic effects for robust antibacterial efficacy. In addition, copper ions liberated from 2A bio-HJs elevate VEGF secretion from fibroblasts, which provokes VEGF receptors (VEGFR) activation through an endogenous pathway, whereas VMP itself promotes an exogenous pathway to facilitate endothelial cell multiplication and tube formation by directly activating the VEGFR signaling pathway. Moreover, employing an in vivo model of infectious ischemic wounds, it is confirmed that the EEB strategy can considerably boost cutaneous regeneration through pathogen elimination, angiogenesis promotion, and collagen deposition. As envisaged, this work leads to the development of a powerful 2A bio-HJ platform that can serve as an effective remedy for bacterial invasion-induced ischemic wounds through the EEB strategy.
Collapse
Affiliation(s)
- Bin Li
- West China Hospital of Stomatology, College of Biomedical Engineering, School of Chemical Engineering, Sichuan University, Chengdu, 610065, China
- State Key Laboratory of Oral Diseases, National Clinical Research Center for Oral Diseases, Department of Orthodontics and Pediatric Dentistry, West China Hospital of Stomatology, Sichuan University, Chengdu, 610065, China
| | - Weizhong Yang
- College of Biomedical Engineering, Sichuan University, Chengdu, 610065, China
| | - Rui Shu
- State Key Laboratory of Oral Diseases, National Clinical Research Center for Oral Diseases, Department of Orthodontics and Pediatric Dentistry, West China Hospital of Stomatology, Sichuan University, Chengdu, 610065, China
| | - Hang Yang
- College of Biomedical Engineering, Sichuan University, Chengdu, 610065, China
| | - Fan Yang
- State Key Laboratory of Oral Diseases, National Clinical Research Center for Oral Diseases, Department of Orthodontics and Pediatric Dentistry, West China Hospital of Stomatology, Sichuan University, Chengdu, 610065, China
| | - Wenyu Dai
- State Key Laboratory of Oral Diseases, National Clinical Research Center for Oral Diseases, Department of Orthodontics and Pediatric Dentistry, West China Hospital of Stomatology, Sichuan University, Chengdu, 610065, China
| | - Wanxi Chen
- State Key Laboratory of Oral Diseases, National Clinical Research Center for Oral Diseases, Department of Orthodontics and Pediatric Dentistry, West China Hospital of Stomatology, Sichuan University, Chengdu, 610065, China
| | - Yau Kei Chan
- Department of Ophthalmology, The University of Hong Kong, Hong Kong, Hong Kong SAR, 999077, China
| | - Ding Bai
- State Key Laboratory of Oral Diseases, National Clinical Research Center for Oral Diseases, Department of Orthodontics and Pediatric Dentistry, West China Hospital of Stomatology, Sichuan University, Chengdu, 610065, China
| | - Yi Deng
- West China Hospital of Stomatology, College of Biomedical Engineering, School of Chemical Engineering, Sichuan University, Chengdu, 610065, China
- State Key Laboratory of Polymer Materials Engineering, Sichuan University, Chengdu, 610065, China
- Department of Mechanical Engineering, The University of Hong Kong, Hong Kong, 999077, China
| |
Collapse
|
204
|
Zhu L, Gou W, Ou L, Liu B, Liu M, Feng H. Role and new insights of microfibrillar-associated protein 4 in fibrotic diseases. APMIS 2024; 132:55-67. [PMID: 37957836 DOI: 10.1111/apm.13358] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2023] [Accepted: 10/24/2023] [Indexed: 11/15/2023]
Abstract
Fibrosis is one of the most worrisome complications of chronic inflammatory diseases, leading to tissue damage, organ failure, and ultimately, death. The most notable pathological characteristic of fibrosis is the excessive accumulation of extracellular matrix (ECM) components such as collagen and fibronectin adjacent to foci of inflammation or damage. The human microfibrillar-associated protein 4 (MFAP4), an important member of the superfamily of fibrinogen-related proteins, is considered to have an extremely important role in ECM transformation of fibrogenesis. This review summarizes the structure, characteristics, and physiological functions of MFAP4 and the importance of MFAP4 in various fibrotic diseases. Meanwhile, we elaborated the underlying actions and mechanisms of MFAP4 in the development of fibrosis, suggesting that a better understand of MFAP4 broadens novel perspective for early screening, diagnosis, prognostic risk assessment, and treatment of fibrotic diseases.
Collapse
Affiliation(s)
- Long Zhu
- Hunan Clinical Research Center of Oral Major Diseases and Oral Health, Changsha, China
- Xiangya Stomatological Hospital, Changsha, China
- Xiangya School of Stomatology, Central South University, Changsha, China
| | - Wenqun Gou
- Hunan Clinical Research Center of Oral Major Diseases and Oral Health, Changsha, China
- Xiangya Stomatological Hospital, Changsha, China
- Xiangya School of Stomatology, Central South University, Changsha, China
- Changsha Stomatological Hospital, Changsha, China
| | - Lijia Ou
- Hunan Clinical Research Center of Oral Major Diseases and Oral Health, Changsha, China
- Department of Histology and Embryology, Xiangya School of Medicine, Central South University, Changsha, China
| | - Binjie Liu
- Hunan Clinical Research Center of Oral Major Diseases and Oral Health, Changsha, China
- Xiangya Stomatological Hospital, Changsha, China
- Xiangya School of Stomatology, Central South University, Changsha, China
| | - Manyi Liu
- Xiangya Stomatological Hospital, Changsha, China
- Xiangya School of Stomatology, Central South University, Changsha, China
| | - Hui Feng
- Hunan Clinical Research Center of Oral Major Diseases and Oral Health, Changsha, China
- Xiangya Stomatological Hospital, Changsha, China
- Xiangya School of Stomatology, Central South University, Changsha, China
| |
Collapse
|
205
|
Bremer J, Pas HH, Diercks GFH, Meijer HJ, van der Molen SM, Nijenhuis AM, van Nijen-Vos LL, Morandé P, Yubero MJ, Palisson F, Fuentes I, Pasmooij AMG. Patients suffering from dystrophic epidermolysis bullosa are prone to developing autoantibodies against skin proteins: A longitudinal confirmational study. Exp Dermatol 2024; 33:e15035. [PMID: 38389191 DOI: 10.1111/exd.15035] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2023] [Revised: 01/22/2024] [Accepted: 02/04/2024] [Indexed: 02/24/2024]
Abstract
Epidermolysis bullosa (EB) is a heritable skin blistering disease caused by variants in genes coding for proteins that secure cell-cell adhesion and attachment of the epidermis to the dermis. Interestingly, several proteins involved in inherited EB are also associated with autoimmune blistering diseases (AIBD). In this study, we present a long-term follow-up of 15 patients suffering from recessive dystrophic or junctional EB. From these patients, 62 sera were analysed for the presence of autoantibodies associated with AIBD. We show that patients suffering from recessive dystrophic EB (RDEB) are more susceptible to developing autoantibodies against skin proteins than patients suffering from junctional EB (70% vs. 20%, respectively). Interestingly, no correlation with age was observed. Most patients showed reactivity to Type XVII collagen/linear IgA bullous dermatosis autoantigen (n = 5; 33%), followed by BP230 (n = 4; 27%), Type VII collagen (n = 4; 27%) and laminin-332 (n = 1; 7%). The pathogenicity of these autoantibodies remains a subject for future experiments.
Collapse
Affiliation(s)
- J Bremer
- Department of Dermatology, Center for Blistering Diseases, University of Groningen, University Medical Center Groningen, Groningen, The Netherlands
| | - H H Pas
- Department of Dermatology, Center for Blistering Diseases, University of Groningen, University Medical Center Groningen, Groningen, The Netherlands
| | - G F H Diercks
- Department of Dermatology, Center for Blistering Diseases, University of Groningen, University Medical Center Groningen, Groningen, The Netherlands
| | - H J Meijer
- Department of Dermatology, Center for Blistering Diseases, University of Groningen, University Medical Center Groningen, Groningen, The Netherlands
| | - S M van der Molen
- Department of Dermatology, Center for Blistering Diseases, University of Groningen, University Medical Center Groningen, Groningen, The Netherlands
| | - A M Nijenhuis
- Department of Dermatology, Center for Blistering Diseases, University of Groningen, University Medical Center Groningen, Groningen, The Netherlands
| | - L L van Nijen-Vos
- Department of Dermatology, Center for Blistering Diseases, University of Groningen, University Medical Center Groningen, Groningen, The Netherlands
| | | | - M J Yubero
- DEBRA Chile, Santiago, Chile
- Pediatrics and Pediatric Infectious Diseases of Clínica Alemana, Facultad de Medicina Alemana, Universidad del Desarrollo, Santiago, Chile
| | - F Palisson
- DEBRA Chile, Santiago, Chile
- Servicio de Dermatología, Facultad de Medicina Clínica Alemana, Universidad del Desarrollo, Santiago, Chile
| | - I Fuentes
- DEBRA Chile, Santiago, Chile
- Centro de Genética y Genómica, Facultad de Medicina Clínica Alemana, Universidad del Desarrollo, Santiago, Chile
- Departamento de Biología Celular y Molecular, Facultad de Ciencias Biológicas, Pontificia Universidad Católica de Chile, Santiago, Chile
| | - A M G Pasmooij
- Department of Dermatology, Center for Blistering Diseases, University of Groningen, University Medical Center Groningen, Groningen, The Netherlands
| |
Collapse
|
206
|
Huang J, Jiang T, Li J, Qie J, Cheng X, Wang Y, Zhou T, Liu J, Han H, Yao K, Yu L. Biomimetic Corneal Stroma for Scarless Corneal Wound Healing via Structural Restoration and Microenvironment Modulation. Adv Healthc Mater 2024; 13:e2302889. [PMID: 37988231 DOI: 10.1002/adhm.202302889] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2023] [Revised: 10/31/2023] [Indexed: 11/23/2023]
Abstract
Corneal injury-induced stromal scarring causes the most common subtype of corneal blindness, and there is an unmet need to promote scarless corneal wound healing. Herein, a biomimetic corneal stroma with immunomodulatory properties is bioengineered for scarless corneal defect repair. First, a fully defined serum-free system is established to derive stromal keratocytes (hAESC-SKs) from a current Good Manufacturing Practice (cGMP)-grade human amniotic epithelial stem cells (hAESCs), and RNA-seq is used to validate the phenotypic transition. Moreover, hAESC-SKs are shown to possess robust immunomodulatory properties in addition to the keratocyte phenotype. Inspired by the corneal stromal extracellular matrix (ECM), a photocurable gelatin-based hydrogel is fabricated to serve as a scaffold for hAESC-SKs for bioengineering of a biomimetic corneal stroma. The rabbit corneal defect model is used to confirm that this biomimetic corneal stroma rapidly restores the corneal structure, and effectively reshapes the tissue microenvironment via proteoglycan secretion to promote transparency and inhibition of the inflammatory cascade to alleviate fibrosis, which synergistically reduces scar formation by ≈75% in addition to promoting wound healing. Overall, the strategy proposed here provides a promising solution for scarless corneal defect repair.
Collapse
Affiliation(s)
- Jianan Huang
- Eye Center, the Second Affiliated Hospital, School of Medicine, Zhejiang University, Zhejiang Provincial Key Laboratory of Ophthalmology, Zhejiang Provincial Clinical Research Center for Eye Diseases, Zhejiang Provincial Engineering Institute on Eye Diseases, Hangzhou, 310009, P. R. China
- MOE Laboratory of Biosystems Homeostasis & Protection & College of Life Sciences-iCell Biotechnology Regenerative Biomedicine Laboratory, College of Life Sciences, Zhejiang University, Hangzhou, 310058, P. R. China
| | - Tuoying Jiang
- MOE Laboratory of Biosystems Homeostasis & Protection & College of Life Sciences-iCell Biotechnology Regenerative Biomedicine Laboratory, College of Life Sciences, Zhejiang University, Hangzhou, 310058, P. R. China
| | - Jinying Li
- MOE Laboratory of Biosystems Homeostasis & Protection & College of Life Sciences-iCell Biotechnology Regenerative Biomedicine Laboratory, College of Life Sciences, Zhejiang University, Hangzhou, 310058, P. R. China
- College of Traditional Chinese Medicine and Health Industry, Lishui University, Lishui, 323000, P. R. China
| | - Jiqiao Qie
- Eye Center, the Second Affiliated Hospital, School of Medicine, Zhejiang University, Zhejiang Provincial Key Laboratory of Ophthalmology, Zhejiang Provincial Clinical Research Center for Eye Diseases, Zhejiang Provincial Engineering Institute on Eye Diseases, Hangzhou, 310009, P. R. China
| | - Xiaoyu Cheng
- Eye Center, the Second Affiliated Hospital, School of Medicine, Zhejiang University, Zhejiang Provincial Key Laboratory of Ophthalmology, Zhejiang Provincial Clinical Research Center for Eye Diseases, Zhejiang Provincial Engineering Institute on Eye Diseases, Hangzhou, 310009, P. R. China
| | - Yiyao Wang
- Eye Center, the Second Affiliated Hospital, School of Medicine, Zhejiang University, Zhejiang Provincial Key Laboratory of Ophthalmology, Zhejiang Provincial Clinical Research Center for Eye Diseases, Zhejiang Provincial Engineering Institute on Eye Diseases, Hangzhou, 310009, P. R. China
| | - Tinglian Zhou
- Eye Center, the Second Affiliated Hospital, School of Medicine, Zhejiang University, Zhejiang Provincial Key Laboratory of Ophthalmology, Zhejiang Provincial Clinical Research Center for Eye Diseases, Zhejiang Provincial Engineering Institute on Eye Diseases, Hangzhou, 310009, P. R. China
| | - Jia Liu
- MOE Laboratory of Biosystems Homeostasis & Protection & College of Life Sciences-iCell Biotechnology Regenerative Biomedicine Laboratory, College of Life Sciences, Zhejiang University, Hangzhou, 310058, P. R. China
| | - Haijie Han
- Eye Center, the Second Affiliated Hospital, School of Medicine, Zhejiang University, Zhejiang Provincial Key Laboratory of Ophthalmology, Zhejiang Provincial Clinical Research Center for Eye Diseases, Zhejiang Provincial Engineering Institute on Eye Diseases, Hangzhou, 310009, P. R. China
- State Key Laboratory of Trauma, Burn and Combined Injury, Third Military Medical University, Chongqing, 400038, P. R. China
| | - Ke Yao
- Eye Center, the Second Affiliated Hospital, School of Medicine, Zhejiang University, Zhejiang Provincial Key Laboratory of Ophthalmology, Zhejiang Provincial Clinical Research Center for Eye Diseases, Zhejiang Provincial Engineering Institute on Eye Diseases, Hangzhou, 310009, P. R. China
| | - Luyang Yu
- MOE Laboratory of Biosystems Homeostasis & Protection & College of Life Sciences-iCell Biotechnology Regenerative Biomedicine Laboratory, College of Life Sciences, Zhejiang University, Hangzhou, 310058, P. R. China
| |
Collapse
|
207
|
Zhou R, Ding RC, Yu Q, Qiu CZ, Zhang HY, Yin ZJ, Ren DL. Metformin Attenuates Neutrophil Recruitment through the H3K18 Lactylation/Reactive Oxygen Species Pathway in Zebrafish. Antioxidants (Basel) 2024; 13:176. [PMID: 38397774 PMCID: PMC10886385 DOI: 10.3390/antiox13020176] [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: 12/15/2023] [Revised: 01/22/2024] [Accepted: 01/25/2024] [Indexed: 02/25/2024] Open
Abstract
Beyond its well-established role in diabetes management, metformin has gained attention as a promising therapeutic for inflammation-related diseases, largely due to its antioxidant capabilities. However, the mechanistic underpinnings of this effect remain elusive. Using in vivo zebrafish models of inflammation, we explored the impact of metformin on neutrophil recruitment and the underlying mechanisms involved. Our data indicate that metformin reduces histone (H3K18) lactylation, leading to the decreased production of reactive oxygen species (ROS) and a muted neutrophil response to both caudal fin injury and otic vesicle inflammation. To investigate the precise mechanisms through which metformin modulates neutrophil migration via ROS and H3K18 lactylation, we meticulously established the correlation between metformin-induced suppression of H3K18 lactylation and ROS levels. Through supplementary experiments involving the restoration of lactate and ROS, our findings demonstrated that elevated levels of both lactate and ROS significantly promoted the inflammatory response in zebrafish. Collectively, our study illuminates previously unexplored avenues of metformin's antioxidant and anti-inflammatory actions through the downregulation of H3K18 lactylation and ROS production, highlighting the crucial role of epigenetic regulation in inflammation and pointing to metformin's potential in treating inflammation-associated conditions.
Collapse
Affiliation(s)
| | | | | | | | | | - Zong-Jun Yin
- College of Animal Science and Technology, Anhui Agricultural University, Hefei 230036, China; (R.Z.); (R.-C.D.); (Q.Y.); (C.-Z.Q.); (H.-Y.Z.)
| | - Da-Long Ren
- College of Animal Science and Technology, Anhui Agricultural University, Hefei 230036, China; (R.Z.); (R.-C.D.); (Q.Y.); (C.-Z.Q.); (H.-Y.Z.)
| |
Collapse
|
208
|
Lin X, Lai Y. Scarring Skin: Mechanisms and Therapies. Int J Mol Sci 2024; 25:1458. [PMID: 38338767 PMCID: PMC10855152 DOI: 10.3390/ijms25031458] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2023] [Revised: 01/19/2024] [Accepted: 01/20/2024] [Indexed: 02/12/2024] Open
Abstract
Skin injury always results in fibrotic, non-functional scars in adults. Although multiple factors are well-known contributors to scar formation, the precise underlying mechanisms remain elusive. This review aims to elucidate the intricacies of the wound healing process, summarize the known factors driving skin cells in wounds toward a scarring fate, and particularly to discuss the impact of fibroblast heterogeneity on scar formation. To the end, we explore potential therapeutic interventions used in the treatment of scarring wounds.
Collapse
Affiliation(s)
- Xinye Lin
- Shanghai Key Laboratory of Regulatory Biology, School of Life Sciences, East China Normal University, Shanghai 200241, China;
- Shanghai Frontiers Science Center of Genome Editing and Cell Therapy, School of Life Sciences, East China Normal University, Shanghai 200241, China
| | - Yuping Lai
- Shanghai Key Laboratory of Regulatory Biology, School of Life Sciences, East China Normal University, Shanghai 200241, China;
- Shanghai Frontiers Science Center of Genome Editing and Cell Therapy, School of Life Sciences, East China Normal University, Shanghai 200241, China
| |
Collapse
|
209
|
Yang J, He Y, Li Z, Yang X, Gao Y, Chen M, Zheng Y, Mao S, Shi X. Intelligent wound dressing for simultaneous in situ detection and elimination of pathogenic bacteria. Acta Biomater 2024; 174:177-190. [PMID: 38070843 DOI: 10.1016/j.actbio.2023.11.045] [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: 08/30/2023] [Revised: 11/19/2023] [Accepted: 11/30/2023] [Indexed: 12/17/2023]
Abstract
Wound infections hinder the healing process and potentially result in life-threatening complications, which urgently require rapid and timely detection and treatment pathogens during the early stages of infection. Here, an intelligent wound dressing was developed to enable in situ detection and elimination of pathogenic bacteria through a combination of point-of-care testing and antibacterial photodynamic therapy technology. The dressing is an injectable hydrogel composed of carboxymethyl chitosan and oxidized sodium alginate, with addition of 4-methylumphulone beta-D-glucoside (MUG) and up-converted nanoparticles coated with titanium dioxide (UCNPs@TiO2). The presence of bacteria can be visually detected by monitoring the blue fluorescence of 4-methylumbellione, generated through the reaction between MUG and the pathogen-associated enzyme. The UCNPs@TiO2 photosensitizers were synthesized and demonstrated high antibacterial activity through the generation of reactive oxygen species when exposed to near-infrared irradiation. Meanwhile, a smartphone-based portable detection system equipped with a self-developed Android app was constructed for in situ detection of pathogens in mere seconds, detecting as few as 103 colony-forming unit. Additionally, the dressing was tested in a rat infected wound model and showed good antibacterial activity and pro-healing ability. These results suggest that the proposed intelligent wound dressing has potential for use in the diagnosis and management of wound infections. STATEMENT OF SIGNIFICANCE: An intelligent wound dressing has been prepared for simultaneous in situ detection and elimination of pathogenic bacteria. The presence of bacteria can be visually detected by tracking the blue fluorescence of the dressing. Moreover, a smartphone-based detection system was constructed to detect and diagnose pathogenic bacteria before reaching the infection limit. Meanwhile, the dressing was able to effectively eliminate key pathogenic bacteria on demand through antibacterial photodynamic therapy under NIR irradiation. The proposed intelligent wound dressing enables timely detection and treatment of infectious pathogens at an early stage, which is beneficial for wound management.
Collapse
Affiliation(s)
- Jianmin Yang
- College of Biological Science and Engineering, Fuzhou University, No. 2 Xueyuan Road, Fuzhou 350108, China; Fujian Key Laboratory of Medical Instrument and Pharmaceutical Technology, Fuzhou University, No. 2 Xueyuan Road, Fuzhou 350108, China; International Joint Laboratory of Intelligent Health Care, Fuzhou University, No. 2 Xueyuan Road, Fuzhou 350108, China
| | - Yuxiang He
- College of Biological Science and Engineering, Fuzhou University, No. 2 Xueyuan Road, Fuzhou 350108, China
| | - Zhendong Li
- College of Biological Science and Engineering, Fuzhou University, No. 2 Xueyuan Road, Fuzhou 350108, China
| | - Xudong Yang
- Fujian Key Laboratory of Medical Instrument and Pharmaceutical Technology, Fuzhou University, No. 2 Xueyuan Road, Fuzhou 350108, China
| | - Yueming Gao
- Fujian Key Laboratory of Medical Instrument and Pharmaceutical Technology, Fuzhou University, No. 2 Xueyuan Road, Fuzhou 350108, China; International Joint Laboratory of Intelligent Health Care, Fuzhou University, No. 2 Xueyuan Road, Fuzhou 350108, China
| | - Mingmao Chen
- College of Biological Science and Engineering, Fuzhou University, No. 2 Xueyuan Road, Fuzhou 350108, China; Fujian Key Laboratory of Medical Instrument and Pharmaceutical Technology, Fuzhou University, No. 2 Xueyuan Road, Fuzhou 350108, China; International Joint Laboratory of Intelligent Health Care, Fuzhou University, No. 2 Xueyuan Road, Fuzhou 350108, China
| | - Yunquan Zheng
- Fujian Key Laboratory of Medical Instrument and Pharmaceutical Technology, Fuzhou University, No. 2 Xueyuan Road, Fuzhou 350108, China; International Joint Laboratory of Intelligent Health Care, Fuzhou University, No. 2 Xueyuan Road, Fuzhou 350108, China
| | - Sifeng Mao
- Department of Applied Chemistry, Graduate School of Urban Environmental Sciences, Tokyo Metropolitan University, 1-1 Minami-Osawa, Hachioji-shi, Tokyo, 192-0397, Japan.
| | - Xianai Shi
- College of Biological Science and Engineering, Fuzhou University, No. 2 Xueyuan Road, Fuzhou 350108, China; Fujian Key Laboratory of Medical Instrument and Pharmaceutical Technology, Fuzhou University, No. 2 Xueyuan Road, Fuzhou 350108, China; International Joint Laboratory of Intelligent Health Care, Fuzhou University, No. 2 Xueyuan Road, Fuzhou 350108, China.
| |
Collapse
|
210
|
Pan Q, Zhang P, Xue F, Zhang J, Fan Z, Chang Z, Liang Z, Zhou G, Ren W. Subcutaneously Engineered Decalcified Bone Matrix Xenografts Promote Bone Repair by Regulating the Immune Microenvironment, Prevascularization, and Stem Cell Homing. ACS Biomater Sci Eng 2024; 10:515-524. [PMID: 38150512 DOI: 10.1021/acsbiomaterials.3c01331] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2023]
Abstract
Immunoregulatory and vascularized microenvironments play an important role in bone regeneration; however, the precise regulation for vascularization and inflammatory reactions remains elusive during bone repair. In this study, by means of subcutaneous preimplantation, we successfully constructed demineralized bone matrix (DBM) grafts with immunoregulatory and vascularized microenvironments. According to the current results, at the early time points (days 1 and 3), subcutaneously implanted DBM grafts recruited a large number of pro-inflammatory M1 macrophages with positive expression of CD68 and iNOS, while at the later time points (days 7 and 14), these inflammatory cells gradually subsided, accompanying increased presence of anti-inflammatory M2 macrophages with positive expression of CD206 and Arg-1, indicating a gradually enhanced anti-inflammatory microenvironment. At the same time, the gradually increased angiogenesis was observed in the DBM grafts with implantation time. In addition, the positive cells of CD105, CD73, and CD90 were observed in the inner region of the DBM grafts, implying the homing of mesenchymal stem cells. The repair results of cranial bone defects in a rat model further confirmed that the subcutaneous DBM xenografts at 7 days significantly improved bone regeneration. In summary, we developed a simple and novel strategy for bone regeneration mediated by anti-inflammatory microenvironment, prevascularization, and endogenous stem cell homing.
Collapse
Affiliation(s)
- Qingqing Pan
- Institutes of Health Central Plain, The Third Affiliated Hospital of Xinxiang Medical University, Clinical Medical Center of Tissue Engineering and Regeneration, Xinxiang Medical University, Xinxiang 453003, China
| | - Pei Zhang
- Institutes of Health Central Plain, The Third Affiliated Hospital of Xinxiang Medical University, Clinical Medical Center of Tissue Engineering and Regeneration, Xinxiang Medical University, Xinxiang 453003, China
| | - Fei Xue
- Institutes of Health Central Plain, The Third Affiliated Hospital of Xinxiang Medical University, Clinical Medical Center of Tissue Engineering and Regeneration, Xinxiang Medical University, Xinxiang 453003, China
| | - Jingxuan Zhang
- Institutes of Health Central Plain, The Third Affiliated Hospital of Xinxiang Medical University, Clinical Medical Center of Tissue Engineering and Regeneration, Xinxiang Medical University, Xinxiang 453003, China
| | - Zhenlin Fan
- Institutes of Health Central Plain, The Third Affiliated Hospital of Xinxiang Medical University, Clinical Medical Center of Tissue Engineering and Regeneration, Xinxiang Medical University, Xinxiang 453003, China
| | - Zhanyu Chang
- Institutes of Health Central Plain, The Third Affiliated Hospital of Xinxiang Medical University, Clinical Medical Center of Tissue Engineering and Regeneration, Xinxiang Medical University, Xinxiang 453003, China
| | - Zhuo Liang
- Institutes of Health Central Plain, The Third Affiliated Hospital of Xinxiang Medical University, Clinical Medical Center of Tissue Engineering and Regeneration, Xinxiang Medical University, Xinxiang 453003, China
| | - Guangdong Zhou
- Department of Plastic and Reconstructive Surgery, Shanghai Ninth People's Hospital, Shanghai Key Laboratory of Tissue Engineering, Shanghai Jiao Tong University School of Medicine, Shanghai 200011, China
| | - Wenjie Ren
- Institutes of Health Central Plain, The Third Affiliated Hospital of Xinxiang Medical University, Clinical Medical Center of Tissue Engineering and Regeneration, Xinxiang Medical University, Xinxiang 453003, China
| |
Collapse
|
211
|
Wu M, Wu S, Tan S, Xu Q, Zhang D, Sun J, Yang H, Wang C, Duan T, Xu Y, Wei Z. VitroGel-loaded human MenSCs promote endometrial regeneration and fertility restoration. Front Bioeng Biotechnol 2024; 11:1310149. [PMID: 38260736 PMCID: PMC10800509 DOI: 10.3389/fbioe.2023.1310149] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2023] [Accepted: 12/15/2023] [Indexed: 01/24/2024] Open
Abstract
Introduction: Intrauterine adhesions (IUA), also known as Asherman's syndrome, is caused by trauma to the pregnant or non-pregnant uterus, which leads to damaged endometrial basal lining and partial or total occlusion of the uterine chambers, resulting in abnormal menstruation, infertility, or recurrent miscarriage. The essence of this syndrome is endometrial fibrosis. And there is no effective treatment for IUA to stimulate endometrial regeneration currently. Recently, menstrual blood-derived stem cells (MenSCs) have been proved to hold therapeutic promise in various diseases, such as myocardial infarction, stroke, diabetes, and liver cirrhosis. Methods: In this study, we examined the effects of MenSCs on the repair of uterine adhesions in a rat model, and more importantly, promoted such therapeutic effects via a xeno-free VitroGel MMP carrier. Results: This combined treatment reduced the expression of inflammatory factors, increased the expression of anti-inflammatory factors, restricted the area of endometrial fibrosis, diminished uterine adhesions, and partially restored fertility, showing stronger effectiveness than each component alone and almost resembling the sham group. Discussion: Our findings suggest a highly promising strategy for IUA treatment.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | | | - Tao Duan
- Shanghai Key Laboratory of Maternal Fetal Medicine, Shanghai Institute of Maternal-Fetal Medicine and Gynecologic Oncology, Clinical and Translational Research Center, Shanghai First Maternity and Infant Hospital, School of Medicine, Tongji University, Shanghai, China
| | - Yao Xu
- Shanghai Key Laboratory of Maternal Fetal Medicine, Shanghai Institute of Maternal-Fetal Medicine and Gynecologic Oncology, Clinical and Translational Research Center, Shanghai First Maternity and Infant Hospital, School of Medicine, Tongji University, Shanghai, China
| | - Zhiyun Wei
- Shanghai Key Laboratory of Maternal Fetal Medicine, Shanghai Institute of Maternal-Fetal Medicine and Gynecologic Oncology, Clinical and Translational Research Center, Shanghai First Maternity and Infant Hospital, School of Medicine, Tongji University, Shanghai, China
| |
Collapse
|
212
|
Anuradha U, Mehra NK, Khatri DK. Understanding molecular mechanisms and miRNA-based targets in diabetes foot ulcers. Mol Biol Rep 2024; 51:82. [PMID: 38183502 DOI: 10.1007/s11033-023-09074-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2023] [Accepted: 11/21/2023] [Indexed: 01/08/2024]
Abstract
In today's culture, obesity and overweight are serious issues that have an impact on how quickly diabetes develops and how it causes complications. For the development of more effective therapies, it is crucial to understand the molecular mechanisms underlying the chronic problems of diabetes. The most prominent effects of diabetes are microvascular abnormalities such as retinopathy, nephropathy, and neuropathy, especially diabetes foot ulcers, as well as macrovascular abnormalities such as heart disease and atherosclerosis. MicroRNAs (miRNAs), which are highly conserved endogenous short non-coding RNA molecules, have been implicated in several physiological functions recently, including the earliest stages of the disease. By binding to particular messenger RNAs (mRNAs), which cause mRNA degradation, translation inhibition, or even gene activation, it primarily regulates posttranscriptional gene expression. These molecules exhibit considerable potential as diagnostic biomarkers for disease and are interesting treatment targets. This review will provide an overview of the latest findings on the key functions that miRNAs role in diabetes and its complications, with an emphasis on the various stages of diabetic wound healing.
Collapse
Affiliation(s)
- Urati Anuradha
- Department of Pharmacology and Toxicology, National Institute of Pharmaceutical Education and Research (NIPER)-Hyderabad, Hyderabad, Telangana, 500037, India
| | - Neelesh Kumar Mehra
- Department of Pharmaceutics, National Institute of Pharmaceutical Education and Research (NIPER)-Hyderabad, Hyderabad, Telangana , 500037, India.
| | - Dharmendra Kumar Khatri
- Department of Pharmacology and Toxicology, National Institute of Pharmaceutical Education and Research (NIPER)-Hyderabad, Hyderabad, Telangana, 500037, India.
| |
Collapse
|
213
|
Wang F, Sun Q, Li Y, Xu R, Li R, Wu D, Huang R, Yang Z, Li Y. Hydrogel Encapsulating Wormwood Essential Oil with Broad-spectrum Antibacterial and Immunomodulatory Properties for Infected Diabetic Wound Healing. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2024; 11:e2305078. [PMID: 38030556 PMCID: PMC10797468 DOI: 10.1002/advs.202305078] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/24/2023] [Revised: 09/29/2023] [Indexed: 12/01/2023]
Abstract
The integration of hydrogels with bio-friendly functional components through simple and efficient strategies to construct wound dressings with broad-spectrum antibacterial and immunomodulatory properties to promote the healing of infected diabetic wounds is highly desirable but remains a major challenge. Here, wormwood essential oil (WEO) is effectively encapsulated in the hydrogel via an O/W-Pickering emulsion during the polymerization of methacrylic anhydride gelatin (GelMA), acrylamide (AM), and acrylic acid N-hydroxysuccinimide ester (AAc-NHS) to form a multifunctional hydrogel dressing (HD-WEO). Compared with conventional emulsions, Pickering emulsions not only improve the encapsulation stability of the WEO, but also enhance the tensile and swelling properties of hydrogel. The synergistic interaction of WEO's diverse bioactive components provides a broad-spectrum antibacterial activity against S. aureus, E. coli, and MRSA. In addition, the HD-WEO can induce the polarization of macrophages from M1 to M2 phenotype. With these advantages, the broad-spectrum antibacterial and immunomodulatory HD-WEO effectively promotes the collagen deposition and neovascularization, thereby accelerating the healing of MRSA-infected diabetic wounds.
Collapse
Affiliation(s)
- Feng Wang
- Guangdong Cardiovascular InstituteGuangdong Provincial People's HospitalGuangdong Academy of Medical SciencesGuangzhou510080China
- Department of Gastrointestinal SurgeryDepartment of General SurgeryGuangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences)Southern Medical UniversityGuangzhou510080China
- Key Laboratory of Biowaste Resources for Selenium‐Enriched Functional Utilization, College of Petroleum and Chemical EngineeringBeibu Gulf UniversityQinzhou535011China
| | - Qi Sun
- School of MedicineSouth China University of TechnologyGuangzhou510006China
| | - Yang Li
- Guangdong Cardiovascular InstituteGuangdong Provincial People's HospitalGuangdong Academy of Medical SciencesGuangzhou510080China
- Department of Gastrointestinal SurgeryDepartment of General SurgeryGuangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences)Southern Medical UniversityGuangzhou510080China
- School of MedicineSouth China University of TechnologyGuangzhou510006China
| | - Ruijun Xu
- School of MedicineSouth China University of TechnologyGuangzhou510006China
| | - Renjie Li
- Guangdong Cardiovascular InstituteGuangdong Provincial People's HospitalGuangdong Academy of Medical SciencesGuangzhou510080China
- Department of Gastrointestinal SurgeryDepartment of General SurgeryGuangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences)Southern Medical UniversityGuangzhou510080China
| | - Dingcai Wu
- PCFM LabSchool of ChemistrySun Yat‐sen UniversityGuangzhou510006China
- The Eighth Affiliated HospitalSun Yat‐sen UniversityShenzhen518033China
| | - Rongkang Huang
- Department of General Surgery (Colorectal Surgery)Guangdong Institute of GastroenterologyBiomedical Innovation CenterGuangdong Provincial Key Laboratory of Colorectal and Pelvic Floor DiseasesThe Sixth Affiliated HospitalSun Yat‐sen UniversityGuangzhou510655China
| | - Zifeng Yang
- Department of Gastrointestinal SurgeryDepartment of General SurgeryGuangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences)Southern Medical UniversityGuangzhou510080China
| | - Yong Li
- Department of Gastrointestinal SurgeryDepartment of General SurgeryGuangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences)Southern Medical UniversityGuangzhou510080China
| |
Collapse
|
214
|
Injarabian L, Willenborg S, Welcker D, Sanin DE, Pasparakis M, Kashkar H, Eming SA. FADD- and RIPK3-Mediated Cell Death Ensures Clearance of Ly6C high Wound Macrophages from Damaged Tissue. J Invest Dermatol 2024; 144:152-164.e7. [PMID: 37516311 DOI: 10.1016/j.jid.2023.06.203] [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: 03/30/2023] [Revised: 06/21/2023] [Accepted: 06/24/2023] [Indexed: 07/31/2023]
Abstract
Cells of the monocyte/macrophage lineage are an integral component of the body's innate ability to restore tissue function after injury. In parallel to mounting an inflammatory response, clearance of monocytes/macrophages from the wound site is critical to re-establish tissue functionality and integrity during the course of healing. The role of regulated cell death in macrophage clearance from damaged tissue and its implications for the outcome of the healing response is little understood. In this study, we explored the role of macrophage-specific FADD-mediated cell death on Ripk3-/- background in a mechanical skin injury model in mice. We found that combined inhibition of RIPK3-mediated necroptosis and FADD-caspase-8-mediated apoptosis in macrophages profoundly delayed wound healing. Importantly, RIPK3 deficiency alone did not considerably alter the wound healing process and macrophage population dynamics, arguing that inhibition of FADD-caspase-8-dependent death of macrophages is primarily responsible for delayed wound closure. Notably, TNF blockade reversed the accumulation of Ly6Chigh macrophages induced by combined deficiency of FADD and RIPK3, indicating a critical dual role of TNF-mediated prosurvival and cell death signaling, particularly in this highly proinflammatory macrophage subset. Our findings reveal a previously uncharacterized cross-talk of inflammatory and cell death signaling in macrophages in regulating repair processes in the skin.
Collapse
Affiliation(s)
| | | | - Daniela Welcker
- Department of Dermatology, University of Cologne, Cologne, Germany
| | - David E Sanin
- Bloomberg-Kimmel Institute for Cancer Immunotherapy, Quantitative Sciences Division and Department of Oncology, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - Manolis Pasparakis
- Institute for Genetics, University of Cologne, Cologne, Germany; Cologne Excellence Cluster on Cellular Stress Responses in Aging-Associated Diseases (CECAD), University of Cologne, Cologne, Germany; Center for Molecular Medicine Cologne (CMMC), University of Cologne, Cologne, Germany
| | - Hamid Kashkar
- Cologne Excellence Cluster on Cellular Stress Responses in Aging-Associated Diseases (CECAD), University of Cologne, Cologne, Germany; Center for Molecular Medicine Cologne (CMMC), University of Cologne, Cologne, Germany; Institute for Molecular Immunology, University of Cologne, Cologne, Germany
| | - Sabine A Eming
- Department of Dermatology, University of Cologne, Cologne, Germany; Cologne Excellence Cluster on Cellular Stress Responses in Aging-Associated Diseases (CECAD), University of Cologne, Cologne, Germany; Center for Molecular Medicine Cologne (CMMC), University of Cologne, Cologne, Germany; Developmental Biology Unit, Institute of Zoology, University of Cologne, Cologne, Germany.
| |
Collapse
|
215
|
Qin Z, Chen Y, Wang Y, Xu Y, Liu T, Mu Q, Huang C. Immunometabolism in the pathogenesis of asthma. Immunology 2024; 171:1-17. [PMID: 37652466 DOI: 10.1111/imm.13688] [Citation(s) in RCA: 17] [Impact Index Per Article: 17.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2023] [Accepted: 08/14/2023] [Indexed: 09/02/2023] Open
Abstract
Bronchial asthma is a heterogeneous disease characterised by chronic airway inflammation. A variety of immune cells such as eosinophils, mast cells, T lymphocytes, neutrophils and airway epithelial cells are involved in the airway inflammation and airway hyperresponsiveness in asthma pathogenesis, resulting in extensive and variable reversible expiratory airflow limitation. However, the precise molecular mechanisms underlying the allergic immune responses, particularly immunometabolism, remains unclear. Studies have detected enhanced oxidative stress, and abnormal metabolic progresses of glycolysis, fatty acid and amino acid in various immune cells, inducing dysregulation of innate and adaptive immune responses in asthma pathogenesis. Immunometabolism mechanisms contain multiple signalling pathways, providing novel therapy targets for asthma. This review summarises the current knowledge on immunometabolism reprogramming in asthma pathogenesis, as well as potential therapy strategies.
Collapse
Affiliation(s)
- Ziwen Qin
- The First Clinical Medical College, Shandong University of Traditional Chinese Medicine, Jinan, Shandong, China
| | - Yujuan Chen
- Experimental Center, Shandong University of Traditional Chinese Medicine, Jinan, Shandong, China
| | - Yue Wang
- The First Clinical Medical College, Shandong University of Traditional Chinese Medicine, Jinan, Shandong, China
| | - Yeyang Xu
- Department of Respiratory and Critical Care Medicine, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, Shandong, China
| | - Tingting Liu
- Department of Respiratory and Critical Care Medicine, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, Shandong, China
| | - Qian Mu
- Department of Respiratory and Critical Care Medicine, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, Shandong, China
| | - Chuanjun Huang
- Department of Respiratory and Critical Care Medicine, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, Shandong, China
| |
Collapse
|
216
|
Wu H, Sun W, Cheng G, Zheng M, Zhao Y, Cao Z. Human Mesenchymal Stem Cells Improve Angiogenesis and Bone Formation in Severed Finger Rats through SIRT1/Nrf2 Signaling. Curr Stem Cell Res Ther 2024; 19:389-399. [PMID: 37183461 DOI: 10.2174/1574888x18666230512112735] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2023] [Revised: 03/15/2023] [Accepted: 03/20/2023] [Indexed: 05/16/2023]
Abstract
BACKGROUND This study employed a severed finger rat model to analyze the effects of human mesenchymal stem cells (MSCs) on angiogenesis, inflammatory response, apoptosis, and oxidative stress, to evaluate the possible mechanism of the repair effect of MSCs on severed finger (SF) rats. METHODS Sixty Sprague-Dawley (SD) rats were categorized into five groups (n = 12). The pathological changes of severed finger tissues were investigated by Hematoxylin and eosin (H&E) staining on day 14 after the rats were sacrificed. The levels of inflammatory factors and oxidative stress factors were detected by ELISA. Terminal Deoxynucleotidyl Transferase (TdT) dUTP Nick End Labeling (TUNEL) was employed to assess the apoptosis of chondrocytes in severed finger tissues. The expression of osteocalcin (OCN), osteopontin (OPN), Collagen I (Col-1), and CD31 were detected by immunohistochemistry or immunofluorescence assay, respectively. The expression levels of related proteins were determined by western blot. RESULT Our study presented evidence that MSCs treatment improved pathological changes of skin and bone tissue, diminished the inflammatory response, prevented oxidative stress injury, suppressed chondrocyte apoptosis, and promoted angiogenesis, and bone formation compared to the model group. In addition, EX527 treatment attenuated the effect of MSCs, SRT1720 and ML385 co-treatment also attenuated the effect of MSCs. Importantly, the MSCs treatment increased the expression of Sirtuin 1(SIRT1)/Nuclear factor erythroid2-related factor 2(Nrf2) relate proteins. CONCLUSION Our study indicated that the mechanism of the effect of MSCs on a severed finger was related to the SIRT1/ Nrf2 signaling pathway.
Collapse
Affiliation(s)
- Hao Wu
- Department of Sports Medicine, Yantaishan Hospital, 264003, Yantai, Shandong, China
| | - Weixue Sun
- Department of Arthrology Surgery, Yantai Yuhuangding Hospital, 264000, Yantai, Shandong, China
| | - Gong Cheng
- Department of Sports Medicine, Yantaishan Hospital, 264003, Yantai, Shandong, China
| | - Mingdi Zheng
- Department of Sports Medicine, Yantaishan Hospital, 264003, Yantai, Shandong, China
| | - Yuchi Zhao
- Department of Articulation Surgery, Yantaishan Hospital, 264003, Yantai, Shandong, China
| | - Zhilin Cao
- Department of Sports Medicine, Yantaishan Hospital, 264003, Yantai, Shandong, China
| |
Collapse
|
217
|
Feng Q, Zhou X, He C. NIR light-facilitated bone tissue engineering. WILEY INTERDISCIPLINARY REVIEWS. NANOMEDICINE AND NANOBIOTECHNOLOGY 2024; 16:e1925. [PMID: 37632228 DOI: 10.1002/wnan.1925] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/17/2023] [Revised: 08/03/2023] [Accepted: 08/05/2023] [Indexed: 08/27/2023]
Abstract
In the last decades, near-infrared (NIR) light has attracted considerable attention due to its unique properties and numerous potential applications in bioimaging and disease treatment. Bone tissue engineering for bone regeneration with the help of biomaterials is currently an effective means of treating bone defects. As a controlled light source with deeper tissue penetration, NIR light can provide real-time feedback of key information on bone regeneration in vivo utilizing fluorescence imaging and be used for bone disease treatment. This review provides a comprehensive overview of NIR light-facilitated bone tissue engineering, from the introduction of NIR probes as well as NIR light-responsive materials, and the visualization of bone regeneration to the treatment of bone-related diseases. Furthermore, the existing challenges and future development directions of NIR light-based bone tissue engineering are also discussed. This article is categorized under: Diagnostic Tools > In Vivo Nanodiagnostics and Imaging Implantable Materials and Surgical Technologies > Nanotechnology in Tissue Repair and Replacement.
Collapse
Affiliation(s)
- Qian Feng
- State Key Laboratory for Modification of Chemical Fibers and Polymer Materials, College of Biological Science and Medical Engineering, Donghua University, Shanghai, China
| | - Xiaojun Zhou
- State Key Laboratory for Modification of Chemical Fibers and Polymer Materials, College of Biological Science and Medical Engineering, Donghua University, Shanghai, China
| | - Chuanglong He
- State Key Laboratory for Modification of Chemical Fibers and Polymer Materials, College of Biological Science and Medical Engineering, Donghua University, Shanghai, China
| |
Collapse
|
218
|
Wei L, Li X, Yao Y, Wang S, Ai X, Liu S. Study on the molecular mechanism of dihydromyricetin in alleviating liver cirrhosis based on network pharmacology. Chem Biol Drug Des 2024; 103:e14421. [PMID: 38230771 DOI: 10.1111/cbdd.14421] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2023] [Revised: 08/31/2023] [Accepted: 12/03/2023] [Indexed: 01/18/2024]
Abstract
Dihydromyricetin (DHM) is a bioactive flavonoid extracted from Hovenia dulcis, which has various activities. In the present study, the molecular mechanism of dihydromyricetin (DHM) in relieving liver cirrhosis was investigated through network pharmacology and experimental verification. The cell model was induced by TGF-β1 activating the human hepatic stellate cell line (HSC; LX-2). The protein levels of α-SMA, collagen I, and collagen III and pathway-related proteins within LX-2 cells were detected using Western blot. EdU staining was conducted to detect cell proliferation. Immunofluorescence staining was performed to detect the expression levels of α-SMA and collagen I. Next, the drug targets of DHM were screened from the PubChem database. The differentially expressed genes in the liver cirrhosis dataset GSE14323 were identified. The expression of the identified drug targets in LX-2 cells was verified using qRT-PCR. The results showed that TGF-β1 treatment notably increased LX-2 cell viability, promoted cell proliferation, and elevated α-SMA, collagen I, and collagen III protein contents. DHM treatment could partially eliminate TGF-β1 effects, as evidenced by the inhibited cell viability and proliferation and reduced α-SMA, collagen I, and collagen III contents. After network pharmacology analysis, nine differentially expressed target genes (MMP2, PDGFRB, PARP1, BCL2L2, ABCB1, TYR, CYP2E1, SQSTM1, and IL6) in liver cirrhosis were identified. According to qRT-PCR verification, DHM could inhibit the expression of MMP2, PDGFRB, PARP1, CYP2E1, SQSTM1, and IL6, and enhance ABCB1 expression levels within LX-2 cells. Moreover, DHM inhibited mTOR and MAPK signaling pathways in TGF-β1-induced HSCs. In conclusion, DHM could inhibit HSC activation, which may be achieved via acting on MMP2, PDGFRB, PARP1, CYP2E1, SQSTM1, IL6, and ABCB1 genes and their downstream signaling pathways, including mTOR and MAPK signaling pathway.
Collapse
Affiliation(s)
- Lin Wei
- College of Basic Medicine, Guizhou University of Traditional Chinese Medicine, Guiyang, Hunan, China
| | - Xiaoying Li
- Key Laboratory of Research and Utilization of Ethnomedicinal Plant Resources of Hunan Province, College of biology and food engineering, Huaihua University, Huaihua, Hunan, China
| | - Yuanzhi Yao
- Key Laboratory of Research and Utilization of Ethnomedicinal Plant Resources of Hunan Province, College of biology and food engineering, Huaihua University, Huaihua, Hunan, China
| | - Siqi Wang
- College of Basic Medicine, Guizhou University of Traditional Chinese Medicine, Guiyang, Hunan, China
| | - Xinghui Ai
- College of Basic Medicine, Guizhou University of Traditional Chinese Medicine, Guiyang, Hunan, China
| | - Shenggui Liu
- Key Laboratory of Research and Utilization of Ethnomedicinal Plant Resources of Hunan Province, College of biology and food engineering, Huaihua University, Huaihua, Hunan, China
| |
Collapse
|
219
|
Choi SB, Kim JH, Kwon S, Ahn NH, Lee JH, Yang WS, Kim CH, Yang SH. 5-methylthiopentyl Isothiocyanate, a Sulforaphane Analogue, Inhibits Pro-inflammatory Cytokines by Regulating LPS/ATP-mediated NLRP3 Inflammasome Activation. Curr Pharm Biotechnol 2024; 25:645-654. [PMID: 37622701 DOI: 10.2174/1389201024666230824093927] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2023] [Revised: 04/25/2023] [Accepted: 05/26/2023] [Indexed: 08/26/2023]
Abstract
BACKGROUND Pro-inflammatory cytokines secreted from activated macrophages and astrocytes are crucial mediators of inflammation for host defense. Among them, the secretion of IL-1β, a major pro-inflammatory cytokine, is especially mediated by the activation of NLRP3 inflammasome. Pro-IL-1β, which is produced in response to the invaded pathogens, such as LPS, is cleaved and matured in the NLRP3 inflammasome by the recognition of ATP. Excessively activated IL-1β induces other immune cells, resulting in the up-regulation of inflammation. Therefore, regulation of NLRP3 inflammasome can be a good strategy for alleviating inflammation. OBJECTIVE Our study aimed to examine whether 5-methylthiopentyl isothiocyanate, a sulforaphane analogue (berteroin), has an anti-inflammatory effect on the NLRP3 inflammasome activation induced by LPS and ATP. METHODS Primary bone marrow-derived macrophages (BMDMs) and astrocytes were stimulated by LPS and ATP with the treatment of 5-methylthiopentyl isothiocyanate, a sulforaphane analogue. The secretion of pro-inflammatory cytokines was measured by ELISA, and the expression level of NLRP3 inflammasome-associated proteins was detected by western blot. The association of NLRP3 inflammasome was assessed by co-immunoprecipitation, and the formation of ASC specks was evaluated by fluorescent microscope. RESULTS 5-methylthiopentyl isothiocyanate, a sulforaphane analogue (berteroin), decreased the release of pro-inflammatory cytokines, IL-1β, and IL-6 in the BMDMs. Berteroin notably prevented the formation of both NLRP3 inflammasome and ASC specks, which reduced the secretion of IL-1β. Additionally, berteroin reduced the IL-1β secretion and cleaved IL-1β expression in the primary astrocytes. DISCUSSION AND CONCLUSION These results indicated the anti-inflammatory effects of 5- methylthiopentyl isothiocyanate (berteroin) by regulating NLRP3 inflammasome activation, suggesting that berteroin could be the potential natural drug candidate for the regulation of inflammation.
Collapse
Affiliation(s)
- Su-Bin Choi
- Department of Biomedical Engineering, College of Life Science and Biotechnology, Dongguk University, Seoul, 04620, Republic of Korea
| | - Ji-Hye Kim
- Department of Biomedical Engineering, College of Life Science and Biotechnology, Dongguk University, Seoul, 04620, Republic of Korea
| | - Sehee Kwon
- Department of Biomedical Engineering, College of Life Science and Biotechnology, Dongguk University, Seoul, 04620, Republic of Korea
| | - Na-Hyun Ahn
- Department of Biomedical Engineering, College of Life Science and Biotechnology, Dongguk University, Seoul, 04620, Republic of Korea
| | - Joo-Hee Lee
- Department of Biomedical Engineering, College of Life Science and Biotechnology, Dongguk University, Seoul, 04620, Republic of Korea
| | - Woong-Suk Yang
- National Institute for Nanomaterials Technology (NINT), POSTECH, 77, Pohang, 37673, Republic of Korea
| | - Cheorl-Ho Kim
- Department of Biological Sciences, Sungkyunkwan University and Samsung Advanced Institute of Health Science and Technology, Suwon, 1649, Republic of Korea
| | - Seung-Hoon Yang
- Department of Biomedical Engineering, College of Life Science and Biotechnology, Dongguk University, Seoul, 04620, Republic of Korea
| |
Collapse
|
220
|
Lee PC, Li CZ, Lu CT, Zhao MH, Lai SM, Liao MH, Peng CL, Liu HT, Lai PS. Microcurrent Cloth-Assisted Transdermal Penetration and Follicular Ducts Escape of Curcumin-Loaded Micelles for Enhanced Wound Healing. Int J Nanomedicine 2023; 18:8077-8097. [PMID: 38164267 PMCID: PMC10758166 DOI: 10.2147/ijn.s440034] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2023] [Accepted: 12/06/2023] [Indexed: 01/03/2024] Open
Abstract
Purpose Larger nanoparticles of bioactive compounds deposit high concentrations in follicular ducts after skin penetration. In this study, we investigated the effects of microcurrent cloth on the skin penetration and translocation of large nanoparticle applied for wound repair applications. Methods A self-assembly of curcumin-loaded micelles (CMs) was prepared to improve the water solubility and transdermal efficiency of curcumin. Microcurrent cloth (M) was produced by Zn/Ag electrofabric printing to facilitate iontophoretic transdermal delivery. The transdermal performance of CMs combined with M was evaluated by a transdermal system and confocal microscopy. The CMs/iontophoretic combination effects on nitric oxide (NO) production and inflammatory cytokines were evaluated in Raw 264.7 cells. The wound-healing property of the combined treatment was assessed in a surgically created full-thickness circular wound mouse model. Results Energy-dispersive X-ray spectroscopy confirmed the presence of Zn/Ag on the microcurrent cloth. The average potential of M was measured to be +214.6 mV in PBS. Large particle CMs (CM-L) prepared using surfactant/cosurfactant present a particle size of 142.9 nm with a polydispersity index of 0.319. The solubility of curcumin in CM-L was 2143.67 μg/mL, indicating 250-fold higher than native curcumin (8.68 μg/mL). The combined treatment (CM-L+M) demonstrated a significant ability to inhibit NO production and increase IL-6 and IL-10 secretion. Surprisingly, microcurrent application significantly improved 20.01-fold transdermal performance of curcumin in CM-L with an obvious escape of CM-L from follicular ducts to surrounding observed by confocal microscopy. The CM-L+M group also exhibited a better wound-closure rate (77.94% on day 4) and the regenerated collagen intensity was approximately 2.66-fold higher than the control group, with a closure rate greater than 90% on day 8 in vivo. Conclusion Microcurrent cloth play as a promising iontophoretic transdermal drug delivery accelerator that enhances skin penetration and assists CMs to escape from follicular ducts for wound repair applications.
Collapse
Affiliation(s)
- Pei-Chi Lee
- xTrans Corporate Research and Innovation Center, Taipei City, Taiwan
| | - Cun-Zhao Li
- Department of Chemistry, National Chung Hsing University, Taichung, Taiwan
| | - Chun-Te Lu
- Division of Plastic and Reconstructive Surgery, Department of Surgery, Taichung Veterans General Hospital, Taichung, Taiwan
- Institute of Medicine, School of Medicine, College of Medicine, National Yang Ming Chiao Tung University, Taipei, Taiwan
| | - Min-Han Zhao
- Department of Chemistry, National Chung Hsing University, Taichung, Taiwan
| | - Syu-Ming Lai
- Department of Chemistry, National Chung Hsing University, Taichung, Taiwan
| | - Man-Hua Liao
- Graduate Institute of Biotechnology, National Chung Hsing University, Taichung, Taiwan
| | - Cheng-Liang Peng
- Isotope Application Division, National Atomic Research Institute, Taoyuan, Taiwan
| | - Hsin-Tung Liu
- xTrans Corporate Research and Innovation Center, Taipei City, Taiwan
| | - Ping-Shan Lai
- Department of Chemistry, National Chung Hsing University, Taichung, Taiwan
| |
Collapse
|
221
|
Dong Q, Fang G, Liu F, Cai S, Tao Y, Xue T, Tang M, Zhang K, An Z, Du J, Zhang H. Ultrasmall calcium-enriched Prussian blue nanozymes promote chronic wound healing by remodeling the wound microenvironment. J Mater Chem B 2023; 11:11578-11587. [PMID: 38014941 DOI: 10.1039/d3tb02065g] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2023]
Abstract
Chronic wound healing remains challenging due to the oxidative microenvironment. Prussian blue (PB) nanoparticles exhibiting multiple antioxidant enzyme-like activities have attracted widespread attention, while their antioxidant efficacy remains unsatisfied. Herein, ultrasmall calcium-enriched Prussian blue nanoparticles (CaPB NPs) are simply constructed with high yields for the wound repair application. Owing to the ultrasmall size and synergistic effect of the generated dual active sites, the CaPB NPs exhibit prominent antioxidase-like activities, protecting cells from oxidative stress-induced damage. In addition to the effect of Ca on regulating keratinocyte and fibroblast growth, it has been demonstrated that the administration of CaPB NPs obviously promoted wound closure as well as collagen deposition and neovascularization in the full-thickness wound defect model in mice. Importantly, the CaPB NP treatment can effectively up-regulate the expression levels of anti-inflammatory cytokines and vascular endothelial growth factors to remodel the wound microenvironment, thereby accelerating the wound healing process. Overall, this work reveals that metal atom substitution is an effective strategy to construct ultrasmall and high-catalytic-performance PB-based nanozymes and further potentiate their effectiveness for chronic wound management.
Collapse
Affiliation(s)
- Qingrong Dong
- Department of Medical Imaging, First Hospital of Shanxi Medical University, Taiyuan 030001, China.
- State Key Laboratory of Radiation Medicine and Protection, School of Radiation Medicine and Protection, School for Radiological and Interdisciplinary Sciences (RAD-X), Collaborative Innovation Center of Radiation Medicine of Jiangsu Higher Education Institutions, Soochow University, Suzhou 215123, China.
- Intelligent Imaging Big Data and Functional Nano-imaging Engineering Research Center of Shanxi Province, Taiyuan 030001, China
| | - Ge Fang
- State Key Laboratory of Radiation Medicine and Protection, School of Radiation Medicine and Protection, School for Radiological and Interdisciplinary Sciences (RAD-X), Collaborative Innovation Center of Radiation Medicine of Jiangsu Higher Education Institutions, Soochow University, Suzhou 215123, China.
| | - Fang Liu
- State Key Laboratory of Radiation Medicine and Protection, School of Radiation Medicine and Protection, School for Radiological and Interdisciplinary Sciences (RAD-X), Collaborative Innovation Center of Radiation Medicine of Jiangsu Higher Education Institutions, Soochow University, Suzhou 215123, China.
- College of Pharmacy, Shanxi Medical University, Jinzhong 030619, China
| | - Shuwei Cai
- State Key Laboratory of Radiation Medicine and Protection, School of Radiation Medicine and Protection, School for Radiological and Interdisciplinary Sciences (RAD-X), Collaborative Innovation Center of Radiation Medicine of Jiangsu Higher Education Institutions, Soochow University, Suzhou 215123, China.
| | - Yujie Tao
- State Key Laboratory of Radiation Medicine and Protection, School of Radiation Medicine and Protection, School for Radiological and Interdisciplinary Sciences (RAD-X), Collaborative Innovation Center of Radiation Medicine of Jiangsu Higher Education Institutions, Soochow University, Suzhou 215123, China.
| | - Tingyu Xue
- Department of Medical Imaging, First Hospital of Shanxi Medical University, Taiyuan 030001, China.
- State Key Laboratory of Radiation Medicine and Protection, School of Radiation Medicine and Protection, School for Radiological and Interdisciplinary Sciences (RAD-X), Collaborative Innovation Center of Radiation Medicine of Jiangsu Higher Education Institutions, Soochow University, Suzhou 215123, China.
- Intelligent Imaging Big Data and Functional Nano-imaging Engineering Research Center of Shanxi Province, Taiyuan 030001, China
| | - Minghua Tang
- Analysis and Testing Center, Soochow University, Suzhou 215123, China
| | - Kun Zhang
- College of Pharmacy, Shanxi Medical University, Jinzhong 030619, China
| | - Ziheng An
- State Key Laboratory of Radiation Medicine and Protection, School of Radiation Medicine and Protection, School for Radiological and Interdisciplinary Sciences (RAD-X), Collaborative Innovation Center of Radiation Medicine of Jiangsu Higher Education Institutions, Soochow University, Suzhou 215123, China.
| | - Jiangfeng Du
- Department of Medical Imaging, First Hospital of Shanxi Medical University, Taiyuan 030001, China.
- Intelligent Imaging Big Data and Functional Nano-imaging Engineering Research Center of Shanxi Province, Taiyuan 030001, China
- College of Pharmacy, Shanxi Medical University, Jinzhong 030619, China
| | - Hui Zhang
- Department of Medical Imaging, First Hospital of Shanxi Medical University, Taiyuan 030001, China.
- Intelligent Imaging Big Data and Functional Nano-imaging Engineering Research Center of Shanxi Province, Taiyuan 030001, China
| |
Collapse
|
222
|
Cai Y, Xiong M, Xin Z, Liu C, Ren J, Yang X, Lei J, Li W, Liu F, Chu Q, Zhang Y, Yin J, Ye Y, Liu D, Fan Y, Sun S, Jing Y, Zhao Q, Zhao L, Che S, Zheng Y, Yan H, Ma S, Wang S, Izpisua Belmonte JC, Qu J, Zhang W, Liu GH. Decoding aging-dependent regenerative decline across tissues at single-cell resolution. Cell Stem Cell 2023; 30:1674-1691.e8. [PMID: 37898124 DOI: 10.1016/j.stem.2023.09.014] [Citation(s) in RCA: 12] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2023] [Revised: 08/28/2023] [Accepted: 09/27/2023] [Indexed: 10/30/2023]
Abstract
Regeneration across tissues and organs exhibits significant variation throughout the body and undergoes a progressive decline with age. To decode the relationships between aging and regenerative capacity, we conducted a comprehensive single-cell transcriptome analysis of regeneration in eight tissues from young and aged mice. We employed diverse analytical models to study tissue regeneration and unveiled the intricate cellular and molecular mechanisms underlying the attenuated regenerative processes observed in aged tissues. Specifically, we identified compromised stem cell mobility and inadequate angiogenesis as prominent contributors to this age-associated decline in regenerative capacity. Moreover, we discovered a unique subset of Arg1+ macrophages that were activated in young tissues but suppressed in aged regenerating tissues, suggesting their important role in age-related immune response disparities during regeneration. This study provides a comprehensive single-cell resource for identifying potential targets for interventions aimed at enhancing regenerative outcomes in the aging population.
Collapse
Affiliation(s)
- Yusheng Cai
- State Key Laboratory of Membrane Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing 100101, China; Institute for Stem Cell and Regeneration, Chinese Academy of Sciences, Beijing 100101, China; Beijing Institute for Stem Cell and Regenerative Medicine, Beijing 100101, China
| | - Muzhao Xiong
- CAS Key Laboratory of Genomic and Precision Medicine, Beijing Institute of Genomics, Chinese Academy of Sciences and China National Center for Bioinformation, Beijing 100101, China; University of Chinese Academy of Sciences, Beijing 100049, China
| | - Zijuan Xin
- State Key Laboratory of Membrane Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing 100101, China; Institute for Stem Cell and Regeneration, Chinese Academy of Sciences, Beijing 100101, China; Beijing Institute for Stem Cell and Regenerative Medicine, Beijing 100101, China
| | - Chengyu Liu
- State Key Laboratory of Stem Cell and Reproductive Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing 100101, China; University of Chinese Academy of Sciences, Beijing 100049, China; Institute for Stem Cell and Regeneration, Chinese Academy of Sciences, Beijing 100101, China; Beijing Institute for Stem Cell and Regenerative Medicine, Beijing 100101, China
| | - Jie Ren
- Key Laboratory of RNA Science and Engineering, Beijing Institute of Genomics, Chinese Academy of Sciences and China National Center for Bioinformation, Beijing 100101, China; CAS Key Laboratory of Genomic and Precision Medicine, Beijing Institute of Genomics, Chinese Academy of Sciences and China National Center for Bioinformation, Beijing 100101, China; Aging Biomarker Consortium, China
| | - Xiying Yang
- Laboratory of Anesthesia and Critical Care Medicine in Colleges and Universities of Shandong Province, School of Anesthesiology, Weifang Medical University, Weifang 261053, China
| | - Jinghui Lei
- Advanced Innovation Center for Human Brain Protection and National Clinical Research Center for Geriatric Disorders, Xuanwu Hospital Capital Medical University, Beijing 100053, China; Aging Translational Medicine Center, International Center for Aging and Cancer, Beijing Municipal Geriatric Medical Research Center, Xuanwu Hospital, Capital Medical University, Beijing 100053, China
| | - Wei Li
- Advanced Innovation Center for Human Brain Protection and National Clinical Research Center for Geriatric Disorders, Xuanwu Hospital Capital Medical University, Beijing 100053, China; Aging Translational Medicine Center, International Center for Aging and Cancer, Beijing Municipal Geriatric Medical Research Center, Xuanwu Hospital, Capital Medical University, Beijing 100053, China
| | - Feifei Liu
- State Key Laboratory of Membrane Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing 100101, China; Institute for Stem Cell and Regeneration, Chinese Academy of Sciences, Beijing 100101, China; Beijing Institute for Stem Cell and Regenerative Medicine, Beijing 100101, China
| | - Qun Chu
- State Key Laboratory of Stem Cell and Reproductive Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing 100101, China; Institute for Stem Cell and Regeneration, Chinese Academy of Sciences, Beijing 100101, China; Beijing Institute for Stem Cell and Regenerative Medicine, Beijing 100101, China
| | - Yiyuan Zhang
- State Key Laboratory of Membrane Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing 100101, China; Institute for Stem Cell and Regeneration, Chinese Academy of Sciences, Beijing 100101, China; Beijing Institute for Stem Cell and Regenerative Medicine, Beijing 100101, China
| | - Jian Yin
- State Key Laboratory of Membrane Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing 100101, China; University of Chinese Academy of Sciences, Beijing 100049, China; Institute for Stem Cell and Regeneration, Chinese Academy of Sciences, Beijing 100101, China; Beijing Institute for Stem Cell and Regenerative Medicine, Beijing 100101, China
| | - Yanxia Ye
- State Key Laboratory of Stem Cell and Reproductive Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing 100101, China; Institute for Stem Cell and Regeneration, Chinese Academy of Sciences, Beijing 100101, China; Beijing Institute for Stem Cell and Regenerative Medicine, Beijing 100101, China
| | - Dingyi Liu
- State Key Laboratory of Stem Cell and Reproductive Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing 100101, China; University of Chinese Academy of Sciences, Beijing 100049, China; Institute for Stem Cell and Regeneration, Chinese Academy of Sciences, Beijing 100101, China; Beijing Institute for Stem Cell and Regenerative Medicine, Beijing 100101, China
| | - Yanling Fan
- CAS Key Laboratory of Genomic and Precision Medicine, Beijing Institute of Genomics, Chinese Academy of Sciences and China National Center for Bioinformation, Beijing 100101, China
| | - Shuhui Sun
- State Key Laboratory of Membrane Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing 100101, China; Institute for Stem Cell and Regeneration, Chinese Academy of Sciences, Beijing 100101, China; Beijing Institute for Stem Cell and Regenerative Medicine, Beijing 100101, China
| | - Yaobin Jing
- State Key Laboratory of Membrane Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing 100101, China; University of Chinese Academy of Sciences, Beijing 100049, China; Institute for Stem Cell and Regeneration, Chinese Academy of Sciences, Beijing 100101, China; Beijing Institute for Stem Cell and Regenerative Medicine, Beijing 100101, China
| | - Qian Zhao
- Advanced Innovation Center for Human Brain Protection and National Clinical Research Center for Geriatric Disorders, Xuanwu Hospital Capital Medical University, Beijing 100053, China; Aging Translational Medicine Center, International Center for Aging and Cancer, Beijing Municipal Geriatric Medical Research Center, Xuanwu Hospital, Capital Medical University, Beijing 100053, China
| | - Liyun Zhao
- Advanced Innovation Center for Human Brain Protection and National Clinical Research Center for Geriatric Disorders, Xuanwu Hospital Capital Medical University, Beijing 100053, China; Aging Translational Medicine Center, International Center for Aging and Cancer, Beijing Municipal Geriatric Medical Research Center, Xuanwu Hospital, Capital Medical University, Beijing 100053, China
| | - Shanshan Che
- CAS Key Laboratory of Genomic and Precision Medicine, Beijing Institute of Genomics, Chinese Academy of Sciences and China National Center for Bioinformation, Beijing 100101, China; University of Chinese Academy of Sciences, Beijing 100049, China
| | - Yandong Zheng
- State Key Laboratory of Stem Cell and Reproductive Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing 100101, China; University of Chinese Academy of Sciences, Beijing 100049, China; Institute for Stem Cell and Regeneration, Chinese Academy of Sciences, Beijing 100101, China; Beijing Institute for Stem Cell and Regenerative Medicine, Beijing 100101, China
| | - Haoteng Yan
- Advanced Innovation Center for Human Brain Protection and National Clinical Research Center for Geriatric Disorders, Xuanwu Hospital Capital Medical University, Beijing 100053, China; Aging Translational Medicine Center, International Center for Aging and Cancer, Beijing Municipal Geriatric Medical Research Center, Xuanwu Hospital, Capital Medical University, Beijing 100053, China
| | - Shuai Ma
- State Key Laboratory of Membrane Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing 100101, China; Institute for Stem Cell and Regeneration, Chinese Academy of Sciences, Beijing 100101, China; Beijing Institute for Stem Cell and Regenerative Medicine, Beijing 100101, China; Aging Biomarker Consortium, China
| | - Si Wang
- Advanced Innovation Center for Human Brain Protection and National Clinical Research Center for Geriatric Disorders, Xuanwu Hospital Capital Medical University, Beijing 100053, China; Aging Translational Medicine Center, International Center for Aging and Cancer, Beijing Municipal Geriatric Medical Research Center, Xuanwu Hospital, Capital Medical University, Beijing 100053, China; Aging Biomarker Consortium, China
| | | | - Jing Qu
- State Key Laboratory of Stem Cell and Reproductive Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing 100101, China; University of Chinese Academy of Sciences, Beijing 100049, China; Institute for Stem Cell and Regeneration, Chinese Academy of Sciences, Beijing 100101, China; Beijing Institute for Stem Cell and Regenerative Medicine, Beijing 100101, China; Aging Biomarker Consortium, China.
| | - Weiqi Zhang
- CAS Key Laboratory of Genomic and Precision Medicine, Beijing Institute of Genomics, Chinese Academy of Sciences and China National Center for Bioinformation, Beijing 100101, China; University of Chinese Academy of Sciences, Beijing 100049, China; Institute for Stem Cell and Regeneration, Chinese Academy of Sciences, Beijing 100101, China; Aging Biomarker Consortium, China.
| | - Guang-Hui Liu
- State Key Laboratory of Membrane Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing 100101, China; University of Chinese Academy of Sciences, Beijing 100049, China; Institute for Stem Cell and Regeneration, Chinese Academy of Sciences, Beijing 100101, China; Beijing Institute for Stem Cell and Regenerative Medicine, Beijing 100101, China; Advanced Innovation Center for Human Brain Protection and National Clinical Research Center for Geriatric Disorders, Xuanwu Hospital Capital Medical University, Beijing 100053, China; Aging Translational Medicine Center, International Center for Aging and Cancer, Beijing Municipal Geriatric Medical Research Center, Xuanwu Hospital, Capital Medical University, Beijing 100053, China; Aging Biomarker Consortium, China.
| |
Collapse
|
223
|
Dou H, Li J, Huang T, Ding X. The sensitivity of mTORC1 signaling activation renders tissue regenerative capacity. CELL REGENERATION (LONDON, ENGLAND) 2023; 12:38. [PMID: 38060073 DOI: 10.1186/s13619-023-00183-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/05/2023] [Accepted: 11/24/2023] [Indexed: 12/08/2023]
Abstract
A better understanding of how and why the regenerative capacity differs among species will not only provide insights into the regeneration process but also hold value for the development of regenerative medicine and the improvement of healing procedures. In a recent Nature article, Zhulyn et al. identify a critical role played by the activation of mechanistic target of rapamycin complex 1 (mTORC1) signaling in enhancing tissue regenerative capacity in animals.
Collapse
Affiliation(s)
- Hanyu Dou
- Institute of Geriatrics, School of Medicine, Affiliated Nantong Hospital of Shanghai University (The Sixth People's Hospital of Nantong), Shanghai University, Nantong, 226011, China
- Shanghai Engineering Research Center of Organ Repair, Shanghai University, Shanghai, 200444, China
| | - Jianzhou Li
- Institute of Geriatrics, School of Medicine, Affiliated Nantong Hospital of Shanghai University (The Sixth People's Hospital of Nantong), Shanghai University, Nantong, 226011, China
- Shanghai Engineering Research Center of Organ Repair, Shanghai University, Shanghai, 200444, China
| | - Taomin Huang
- Department of Pharmacy, Eye & ENT Hospital, Fudan University, Shanghai, 200031, China.
| | - Xiaolei Ding
- Institute of Geriatrics, School of Medicine, Affiliated Nantong Hospital of Shanghai University (The Sixth People's Hospital of Nantong), Shanghai University, Nantong, 226011, China.
- Shanghai Engineering Research Center of Organ Repair, Shanghai University, Shanghai, 200444, China.
- Department of Dermatology, Huashan Hospital, Fudan University, Shanghai, China.
| |
Collapse
|
224
|
Maldonado H, Savage BD, Barker HR, May U, Vähätupa M, Badiani RK, Wolanska KI, Turner CMJ, Pemmari T, Ketomäki T, Prince S, Humphries MJ, Ruoslahti E, Morgan MR, Järvinen TAH. Systemically administered wound-homing peptide accelerates wound healing by modulating syndecan-4 function. Nat Commun 2023; 14:8069. [PMID: 38057316 PMCID: PMC10700342 DOI: 10.1038/s41467-023-43848-1] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2022] [Accepted: 11/19/2023] [Indexed: 12/08/2023] Open
Abstract
CAR (CARSKNKDC) is a wound-homing peptide that recognises angiogenic neovessels. Here we discover that systemically administered CAR peptide has inherent ability to promote wound healing: wounds close and re-epithelialise faster in CAR-treated male mice. CAR promotes keratinocyte migration in vitro. The heparan sulfate proteoglycan syndecan-4 regulates cell migration and is crucial for wound healing. We report that syndecan-4 expression is restricted to epidermis and blood vessels in mice skin wounds. Syndecan-4 regulates binding and internalisation of CAR peptide and CAR-mediated cytoskeletal remodelling. CAR induces syndecan-4-dependent activation of the small GTPase ARF6, via the guanine nucleotide exchange factor cytohesin-2, and promotes syndecan-4-, ARF6- and Cytohesin-2-mediated keratinocyte migration. Finally, we show that genetic ablation of syndecan-4 in male mice eliminates CAR-induced wound re-epithelialisation following systemic administration. We propose that CAR peptide activates syndecan-4 functions to selectively promote re-epithelialisation. Thus, CAR peptide provides a therapeutic approach to enhance wound healing in mice; systemic, yet target organ- and cell-specific.
Collapse
Affiliation(s)
- Horacio Maldonado
- Institute of Systems, Molecular & Integrative Biology, University of Liverpool, Liverpool, UK
| | - Bryan D Savage
- Institute of Systems, Molecular & Integrative Biology, University of Liverpool, Liverpool, UK
| | - Harlan R Barker
- Faculty of Medicine and Health Technology, Tampere University & Tampere University Hospital, Tampere, Finland
| | - Ulrike May
- Faculty of Medicine and Health Technology, Tampere University & Tampere University Hospital, Tampere, Finland
| | - Maria Vähätupa
- Faculty of Medicine and Health Technology, Tampere University & Tampere University Hospital, Tampere, Finland
| | - Rahul K Badiani
- Institute of Systems, Molecular & Integrative Biology, University of Liverpool, Liverpool, UK
| | - Katarzyna I Wolanska
- Institute of Systems, Molecular & Integrative Biology, University of Liverpool, Liverpool, UK
| | - Craig M J Turner
- Institute of Systems, Molecular & Integrative Biology, University of Liverpool, Liverpool, UK
| | - Toini Pemmari
- Faculty of Medicine and Health Technology, Tampere University & Tampere University Hospital, Tampere, Finland
| | - Tuomo Ketomäki
- Faculty of Medicine and Health Technology, Tampere University & Tampere University Hospital, Tampere, Finland
| | - Stuart Prince
- Faculty of Medicine and Health Technology, Tampere University & Tampere University Hospital, Tampere, Finland
| | - Martin J Humphries
- Wellcome Trust Centre for Cell-Matrix Research, University of Manchester, Manchester, UK
| | - Erkki Ruoslahti
- Cancer Center, Sanford Burnham Prebys Medical Discovery Institute, La Jolla, CA and Center for Nanomedicine, University of California (UCSB), Santa Barbara, CA, USA
| | - Mark R Morgan
- Institute of Systems, Molecular & Integrative Biology, University of Liverpool, Liverpool, UK.
| | - Tero A H Järvinen
- Faculty of Medicine and Health Technology, Tampere University & Tampere University Hospital, Tampere, Finland.
- Cancer Center, Sanford Burnham Prebys Medical Discovery Institute, La Jolla, CA and Center for Nanomedicine, University of California (UCSB), Santa Barbara, CA, USA.
| |
Collapse
|
225
|
Casari M, Siegl D, Deppermann C, Schuppan D. Macrophages and platelets in liver fibrosis and hepatocellular carcinoma. Front Immunol 2023; 14:1277808. [PMID: 38116017 PMCID: PMC10728659 DOI: 10.3389/fimmu.2023.1277808] [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: 08/15/2023] [Accepted: 11/13/2023] [Indexed: 12/21/2023] Open
Abstract
During fibrosis, (myo)fibroblasts deposit large amounts of extracellular matrix proteins, thereby replacing healthy functional tissue. In liver fibrosis, this leads to the loss of hepatocyte function, portal hypertension, variceal bleeding, and increased susceptibility to infection. At an early stage, liver fibrosis is a dynamic and reversible process, however, from the cirrhotic stage, there is significant progression to hepatocellular carcinoma. Both liver-resident macrophages (Kupffer cells) and monocyte-derived macrophages are important drivers of fibrosis progression, but can also induce its regression once triggers of chronic inflammation are eliminated. In liver cancer, they are attracted to the tumor site to become tumor-associated macrophages (TAMs) polarized towards a M2- anti-inflammatory/tumor-promoting phenotype. Besides their role in thrombosis and hemostasis, platelets can also stimulate fibrosis and tumor development by secreting profibrogenic factors and regulating the innate immune response, e.g., by interacting with monocytes and macrophages. Here, we review recent literature on the role of macrophages and platelets and their interplay in liver fibrosis and hepatocellular carcinoma.
Collapse
Affiliation(s)
- Martina Casari
- Center for Thrombosis and Hemostasis, University Medical Center of the Johannes Gutenberg-University, Mainz, Germany
| | - Dominik Siegl
- Institute for Translational Immunology, University Medical Center of the Johannes Gutenberg-University, Mainz, Germany
| | - Carsten Deppermann
- Center for Thrombosis and Hemostasis, University Medical Center of the Johannes Gutenberg-University, Mainz, Germany
- Research Center for Immune Therapy Forschungszentrum für Immuntherapie (FZI), University Medical Center of the Johannes Gutenberg-University, Mainz, Germany
| | - Detlef Schuppan
- Institute for Translational Immunology, University Medical Center of the Johannes Gutenberg-University, Mainz, Germany
- Research Center for Immune Therapy Forschungszentrum für Immuntherapie (FZI), University Medical Center of the Johannes Gutenberg-University, Mainz, Germany
- Division of Gastroenterology, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA, United States
| |
Collapse
|
226
|
Cappitti A, Palmieri F, Garella R, Tani A, Chellini F, Salzano De Luna M, Parmeggiani C, Squecco R, Martella D, Sassoli C. Development of accessible platforms to promote myofibroblast differentiation by playing on hydrogel scaffold composition. BIOMATERIALS ADVANCES 2023; 155:213674. [PMID: 37922662 DOI: 10.1016/j.bioadv.2023.213674] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/11/2023] [Revised: 10/19/2023] [Accepted: 10/26/2023] [Indexed: 11/07/2023]
Abstract
Mechanomimetic materials are particularly attractive for modeling in vitro fibroblast to myofibroblast (Myof) transition, a key process in the physiological repair of damaged tissue, and recognized as the core cellular mechanism of pathological fibrosis in different organs. In vivo, mechanical stimuli from the extracellular matrix (ECM) are crucial, together with cell-cell contacts and the pro-fibrotic transforming growth factor (TGF)-β1, in promoting fibroblast differentiation. Here, we explore the impact of hydrogels made by polyacrylamide with different composition on fibroblast behavior. By appropriate modulation of the hydrogel composition (e.g. adjusting the crosslinker content), we produce and fully characterize three kinds of scaffolds with different Young modulus (E). We observe that soft hydrogels (E < 1 kPa) induced fibroblast differentiation better than stiffer ones, also in the absence of TGF-β1. This study provides a readily accessible biomaterial platform to promote Myof generation. The easy approach used and the commercial availability of the monomers make these hydrogels suitable to a wide range of biomedical applications combined with high reproducibility and simple preparation protocols.
Collapse
Affiliation(s)
- Alice Cappitti
- Department of Chemistry "Ugo Schiff", University of Florence, 50019 Sesto Fiorentino, Italy
| | - Francesco Palmieri
- Department of Experimental and Clinical Medicine, Section of Physiological Sciences, University of Florence, 50134 Florence, Italy
| | - Rachele Garella
- Department of Experimental and Clinical Medicine, Section of Physiological Sciences, University of Florence, 50134 Florence, Italy
| | - Alessia Tani
- Department of Experimental and Clinical Medicine, Section of Anatomy and Histology, Imaging Platform, University of Florence, 50134 Florence, Italy
| | - Flaminia Chellini
- Department of Experimental and Clinical Medicine, Section of Anatomy and Histology, Imaging Platform, University of Florence, 50134 Florence, Italy
| | - Martina Salzano De Luna
- Department of chemical, materials and industrial production engineering, University of Naples Federico II, 80125 Napoli, Italy
| | - Camilla Parmeggiani
- Department of Chemistry "Ugo Schiff", University of Florence, 50019 Sesto Fiorentino, Italy; European Laboratory for Non-Linear Spectroscopy (LENS), 50019 Sesto Fiorentino, Italy
| | - Roberta Squecco
- Department of Experimental and Clinical Medicine, Section of Physiological Sciences, University of Florence, 50134 Florence, Italy.
| | - Daniele Martella
- Department of Chemistry "Ugo Schiff", University of Florence, 50019 Sesto Fiorentino, Italy; European Laboratory for Non-Linear Spectroscopy (LENS), 50019 Sesto Fiorentino, Italy; Istituto Nazionale di Ricerca Metrologica (INRiM), 10135 Torino, Italy.
| | - Chiara Sassoli
- Department of Experimental and Clinical Medicine, Section of Anatomy and Histology, Imaging Platform, University of Florence, 50134 Florence, Italy
| |
Collapse
|
227
|
Monaghan MG, Borah R, Thomsen C, Browne S. Thou shall not heal: Overcoming the non-healing behaviour of diabetic foot ulcers by engineering the inflammatory microenvironment. Adv Drug Deliv Rev 2023; 203:115120. [PMID: 37884128 DOI: 10.1016/j.addr.2023.115120] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2023] [Revised: 08/01/2023] [Accepted: 10/23/2023] [Indexed: 10/28/2023]
Abstract
Diabetic foot ulcers (DFUs) are a devastating complication for diabetic patients that have debilitating effects and can ultimately lead to limb amputation. Healthy wounds progress through the phases of healing leading to tissue regeneration and restoration of the barrier function of the skin. In contrast, in diabetic patients dysregulation of these phases leads to chronic, non-healing wounds. In particular, unresolved inflammation in the DFU microenvironment has been identified as a key facet of chronic wounds in hyperglyceamic patients, as DFUs fail to progress beyond the inflammatory phase and towards resolution. Thus, control over and modulation of the inflammatory response is a promising therapeutic avenue for DFU treatment. This review discusses the current state-of-the art regarding control of the inflammatory response in the DFU microenvironment, with a specific focus on the development of biomaterials-based delivery strategies and their cargos to direct tissue regeneration in the DFU microenvironment.
Collapse
Affiliation(s)
- Michael G Monaghan
- Department of Mechanical, Manufacturing and Biomedical Engineering, Trinity College Dublin, Dublin 2, Ireland; Advanced Materials and BioEngineering Research (AMBER), Centre at Trinity College Dublin and the Royal College of Surgeons in Ireland, Dublin 2, Ireland; CÚRAM, Centre for Research in Medical Devices, National University of Ireland, H91 W2TY Galway, Ireland; Trinity Centre for Biomedical Engineering, Trinity College Dublin, Dublin 2, Ireland
| | - Rajiv Borah
- Department of Mechanical, Manufacturing and Biomedical Engineering, Trinity College Dublin, Dublin 2, Ireland; Advanced Materials and BioEngineering Research (AMBER), Centre at Trinity College Dublin and the Royal College of Surgeons in Ireland, Dublin 2, Ireland; Trinity Centre for Biomedical Engineering, Trinity College Dublin, Dublin 2, Ireland
| | - Charlotte Thomsen
- Department of Mechanical, Manufacturing and Biomedical Engineering, Trinity College Dublin, Dublin 2, Ireland; Tissue Engineering Research Group, Department of Anatomy and Regenerative Medicine, Royal College of Surgeons in Ireland, Dublin, Ireland
| | - Shane Browne
- CÚRAM, Centre for Research in Medical Devices, National University of Ireland, H91 W2TY Galway, Ireland; Trinity Centre for Biomedical Engineering, Trinity College Dublin, Dublin 2, Ireland; Tissue Engineering Research Group, Department of Anatomy and Regenerative Medicine, Royal College of Surgeons in Ireland, Dublin, Ireland.
| |
Collapse
|
228
|
Tehrany PM, Rahmanian P, Rezaee A, Ranjbarpazuki G, Sohrabi Fard F, Asadollah Salmanpour Y, Zandieh MA, Ranjbarpazuki A, Asghari S, Javani N, Nabavi N, Aref AR, Hashemi M, Rashidi M, Taheriazam A, Motahari A, Hushmandi K. Multifunctional and theranostic hydrogels for wound healing acceleration: An emphasis on diabetic-related chronic wounds. ENVIRONMENTAL RESEARCH 2023; 238:117087. [PMID: 37716390 DOI: 10.1016/j.envres.2023.117087] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/16/2023] [Revised: 08/28/2023] [Accepted: 09/05/2023] [Indexed: 09/18/2023]
Abstract
Hydrogels represent intricate three-dimensional polymeric structures, renowned for their compatibility with living systems and their ability to naturally degrade. These networks stand as promising and viable foundations for a range of biomedical uses. The practical feasibility of employing hydrogels in clinical trials has been well-demonstrated. Among the prevalent biomedical uses of hydrogels, a significant application arises in the context of wound healing. This intricate progression involves distinct phases of inflammation, proliferation, and remodeling, often triggered by trauma, skin injuries, and various diseases. Metabolic conditions like diabetes have the potential to give rise to persistent wounds, leading to delayed healing processes. This current review consolidates a collection of experiments focused on the utilization of hydrogels to expedite the recovery of wounds. Hydrogels have the capacity to improve the inflammatory conditions at the wound site, and they achieve this by diminishing levels of reactive oxygen species (ROS), thereby exhibiting antioxidant effects. Hydrogels have the potential to enhance the growth of fibroblasts and keratinocytes at the wound site. They also possess the capability to inhibit both Gram-positive and Gram-negative bacteria, effectively managing wounds infected by drug-resistant bacteria. Hydrogels can trigger angiogenesis and neovascularization processes, while also promoting the M2 polarization of macrophages, which in turn mitigates inflammation at the wound site. Intelligent and versatile hydrogels, encompassing features such as pH sensitivity, reactivity to reactive oxygen species (ROS), and responsiveness to light and temperature, have proven advantageous in expediting wound healing. Furthermore, hydrogels synthesized using environmentally friendly methods, characterized by high levels of biocompatibility and biodegradability, hold the potential for enhancing the wound healing process. Hydrogels can facilitate the controlled discharge of bioactive substances. More recently, there has been progress in the creation of conductive hydrogels, which, when subjected to electrical stimulation, contribute to the enhancement of wound healing. Diabetes mellitus, a metabolic disorder, leads to a slowdown in the wound healing process, often resulting in the formation of persistent wounds. Hydrogels have the capability to expedite the healing of diabetic wounds, facilitating the transition from the inflammatory phase to the proliferative stage. The current review sheds light on the biological functionalities of hydrogels, encompassing their role in modulating diverse mechanisms and cell types, including inflammation, oxidative stress, macrophages, and bacteriology. Additionally, this review emphasizes the significance of smart hydrogels with responsiveness to external stimuli, as well as conductive hydrogels for promoting wound healing. Lastly, the discussion delves into the advancement of environmentally friendly hydrogels with high biocompatibility, aimed at accelerating the wound healing process.
Collapse
Affiliation(s)
| | - Parham Rahmanian
- Faculty of Veterinary Medicine, Islamic Azad University, Science and Research Branch, Tehran, Iran
| | - Aryan Rezaee
- Student Research Committee, School of Medicine, Iran University of Medical Sciences, Tehran, Iran
| | - Golnaz Ranjbarpazuki
- Student Research Committee, School of Medicine, Iran University of Medical Sciences, Tehran, Iran
| | - Farima Sohrabi Fard
- Faculty of Veterinary Medicine, Islamic Azad University, Science and Research Branch, Tehran, Iran
| | | | - Mohammad Arad Zandieh
- Department of Food Hygiene and Quality Control, Division of Epidemiology, Faculty of Veterinary Medicine, University of Tehran, Tehran, Iran
| | - Ali Ranjbarpazuki
- Student Research Committee, School of Medicine, Iran University of Medical Sciences, Tehran, Iran
| | - Sajedeh Asghari
- Faculty of Veterinary Medicine, Islamic Azad University, Babol Branch, Babol, Iran
| | - Nazanin Javani
- Department of Food Science and Technology, Faculty of Pharmacy, Tehran Medical Sciences, Islamic Azad University, Tehran, Iran
| | - Noushin Nabavi
- Department of Urologic Sciences, University of British Columbia, Vancouver, BC, V5Z 1M9, Canada
| | - Amir Reza Aref
- Belfer Center for Applied Cancer Science, Dana-Farber Cancer Institute, Harvard Medical School, Boston, MA, USA; Department of Translational Sciences, Xsphera Biosciences Inc. Boston, MA, USA
| | - Mehrdad Hashemi
- Department of Genetics, Faculty of Advanced Science and Technology, Tehran Medical Sciences, Islamic Azad University, Tehran, Iran; Farhikhtegan Medical Convergence Sciences Research Center, Farhikhtegan Hospital Tehran Medical Sciences, Islamic Azad University, Tehran, Iran
| | - Mohsen Rashidi
- Department Pharmacology, Faculty of Medicine, Mazandaran University of Medical Sciences, Sari, Iran; The Health of Plant and Livestock Products Research Center, Mazandaran University of Medical Sciences, Sari, Iran.
| | - Afshin Taheriazam
- Department of Genetics, Faculty of Advanced Science and Technology, Tehran Medical Sciences, Islamic Azad University, Tehran, Iran; Department of Orthopedics, Faculty of Medicine, Tehran Medical Sciences, Islamic Azad University, Tehran, Iran.
| | - Alireza Motahari
- Board-Certified in Veterinary Surgery, School of Veterinary Medicine, Shiraz University, Shiraz, Iran.
| | - Kiavash Hushmandi
- Department of Food Hygiene and Quality Control, Division of Epidemiology, Faculty of Veterinary Medicine, University of Tehran, Tehran, Iran.
| |
Collapse
|
229
|
Satzinger S, Willenborg S, Ding X, Klose CSN, Radtke D, Voehringer D, Eming SA. Type 2 Immunity Regulates Dermal White Adipose Tissue Function. J Invest Dermatol 2023; 143:2456-2467.e5. [PMID: 37295491 DOI: 10.1016/j.jid.2023.05.017] [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: 05/13/2022] [Revised: 04/19/2023] [Accepted: 05/09/2023] [Indexed: 06/12/2023]
Abstract
Type 2 immune responses have been increasingly linked with tissue maintenance, regeneration, and metabolic homeostasis. The molecular basis of regulator and effector mechanisms of type 2 immunity in skin regeneration and homeostasis is still lacking. In this study, we analyzed the role of IL-4Rα signaling in the regeneration of diverse cellular compartments in the skin. Mutants with global IL-4Rα deficiency showed two major phenotypes: first, a pronounced atrophy of the interfollicular epidermis, and second, a significant increase in dermal white adipose tissue thickness in mice aged 3 weeks (postnatal day 21) compared with littermate controls. Notably, IL-4Rα deficiency decreased the activation of hormone-sensitive lipase, a rate-limiting step in lipolysis. Immunohistochemical and FACS analysis in IL-4/enhanced GFP reporter mice showed that IL-4 expression peaked on postnatal day 21 and that eosinophils are the predominant IL-4-expressing cells. Eosinophil-deficient mice recapitulated the lipolytic-defective dermal white adipose tissue phenotype of Il4ra-deficient mice, showing that eosinophils are necessary for dermal white adipose tissue lipolysis. Collectively, we provide mechanistic insights into the regulation of interfollicular epidermis and hormone-sensitive lipase-mediated lipolysis in dermal white adipose tissue in early life by IL-4Rα, and our findings show that eosinophils play a critical role in this process.
Collapse
Affiliation(s)
| | | | - Xiaolei Ding
- Department of Dermatology, University of Cologne, Cologne, Germany; Institute of Geriatrics (Shanghai University), Affiliated Nantong Hospital of Shanghai University (The Sixth People's Hospital of Nantong), School of Medicine, Shanghai University, Nantong, China; Shanghai Engineering Research Center of Organ Repair, School of Medicine, Shanghai University, Shanghai, China
| | - Christoph S N Klose
- Department of Microbiology, Infectious Diseases and Immunology, Charité - Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Berlin, Germany
| | - Daniel Radtke
- Department of Infection Biology, University Hospital Erlangen and Friedrich-Alexander Universität Erlangen-Nürnberg (FAU), Erlangen, Germany
| | - David Voehringer
- Department of Infection Biology, University Hospital Erlangen and Friedrich-Alexander Universität Erlangen-Nürnberg (FAU), Erlangen, Germany
| | - Sabine A Eming
- Department of Dermatology, University of Cologne, Cologne, Germany; Center for Molecular Medicine Cologne (CMMC), University of Cologne, Cologne, Germany; Cologne Excellence Cluster on Cellular Stress Responses in Aging-Associated Diseases (CECAD), University of Cologne, Cologne, Germany; Institute of Zoology, Developmental Biology Unit, University of Cologne, Cologne, Germany.
| |
Collapse
|
230
|
Xu Z, Cheng C, Zhang Y, Yang D, Jing W, Liu X, Li X. Lipopolysaccharide induces skin scarring through the TLR4/Myd88 inflammatory signaling pathway in dermal fibroblasts. Burns 2023; 49:1997-2006. [PMID: 37821278 DOI: 10.1016/j.burns.2023.06.005] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2022] [Revised: 04/19/2023] [Accepted: 06/12/2023] [Indexed: 10/13/2023]
Abstract
Skin scarring is a frequent complication of the wound healing process. Bacterial contamination and prolonged inflammation in wounds are thought to play significant roles during scar formation, but little is known about their specific mechanisms of action. In this study, hypertrophic scar derived fibroblasts (HSFs) and paired normal skin derived fibroblasts (NSFs) were used to evaluate the effects of lipopolysaccharide (LPS) on inflammation-induced skin scarring and explore the inflammation-mediated mechanism of activity of LPS on dermal fibroblasts. LPS was found to significantly upregulate the expression of the proinflammatory molecules TLR4, Myd88, TRAF6, and p65, and the fibrosis-related proteins Col I, Col III, and α-SMA, in NSFs. Blocking Myd88 expression with T6167923 downregulated the expression of Col I, Col III, and α-SMA, whereas activating Myd88 expression with CL075 significantly upregulated their expression in LPS-treated NSFs. LPS was found to delay wound healing and increase skin scarring in cell and mouse models. These results showed that LPS could induce scar formation through the TLR4/Myd88 signaling pathway in dermal fibroblasts, suggesting that the downregulation of excessive inflammation in wound tissues inhibits skin scarring and improves scar appearance.
Collapse
Affiliation(s)
- Zhigang Xu
- Department of Dermatology, the Second Affiliated Hospital of Xi'an Jiaotong University, Xi Wu Road, No 157, Xi'an 710004, China; Department of Burns and Cutaneous Surgery, Xijing Hospital, Air Force Medical University, West Chang-le Road, No 127, Xi'an 710032, China
| | - Chuantao Cheng
- Department of Dermatology, the Second Affiliated Hospital of Xi'an Jiaotong University, Xi Wu Road, No 157, Xi'an 710004, China
| | - Yangang Zhang
- Department of Dermatology, the Second Affiliated Hospital of Xi'an Jiaotong University, Xi Wu Road, No 157, Xi'an 710004, China
| | - Danyang Yang
- Department of Dermatology, the Second Affiliated Hospital of Xi'an Jiaotong University, Xi Wu Road, No 157, Xi'an 710004, China
| | - Wenwen Jing
- Department of Dermatology, the Second Affiliated Hospital of Xi'an Jiaotong University, Xi Wu Road, No 157, Xi'an 710004, China
| | - Xin Liu
- Department of Dermatology, the Second Affiliated Hospital of Xi'an Jiaotong University, Xi Wu Road, No 157, Xi'an 710004, China
| | - Xiaoli Li
- Department of Dermatology, the Second Affiliated Hospital of Xi'an Jiaotong University, Xi Wu Road, No 157, Xi'an 710004, China.
| |
Collapse
|
231
|
Abstract
Tissue regeneration is not simply a local repair event occurring in isolation from the distant, uninjured parts of the body. Rather, evidence indicates that regeneration is a whole-animal process involving coordinated interactions between different organ systems. Here, we review recent studies that reveal how remote uninjured tissues and organ systems respond to and engage in regeneration. We also discuss the need for toolkits and technological advancements to uncover and dissect organ communication during regeneration.
Collapse
Affiliation(s)
- Fei Sun
- Duke Regeneration Center, Department of Cell Biology, Duke University School of Medicine, Durham, NC 27710, USA
| | - Kenneth D. Poss
- Duke Regeneration Center, Department of Cell Biology, Duke University School of Medicine, Durham, NC 27710, USA
| |
Collapse
|
232
|
Huang Y, Zhang Q, Cao W, Zhang Q, Wang L, Lai D. TNF-α and IFN-γ prestimulation enhances the therapeutic efficacy of human amniotic epithelial stem cells in chemotherapy-induced ovarian dysfunction. Inflamm Regen 2023; 43:57. [PMID: 37993924 PMCID: PMC10664537 DOI: 10.1186/s41232-023-00309-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/04/2023] [Accepted: 11/11/2023] [Indexed: 11/24/2023] Open
Abstract
BACKGROUND Exposure to a harsh ovarian microenvironment induced by chemotherapeutic agents seriously affects the remodeling of ovarian function and follicular development, leading to premature ovarian failure or insufficiency (POF/POI). For decades, the effectiveness of stem cell therapies in POI animal models has been intensively studied; however, strategies to enhance the therapeutic effect of stem cells remain challenging. METHODS In this study, we first observed the pathological changes of the ovaries at different time points during chemotherapy, including the number of follicles, granulosa cell proliferation, oxidative stress damage, ovarian fibrosis, and inflammatory reaction. Moreover, we investigated whether activated hAECs stimulated by the proinflammatory cytokines tumor necrosis factor-α (TNF-α) and interferon-γ (IFN-γ) were more effective than native hAECs in repairing ovarian injury induced by chemotherapy. RESULTS The inhibitory effect of chemotherapy drugs on ovarian granulosa cells (GCs) in growing follicles mainly occurred on day 3 after chemotherapy in a mouse model. Then, continued ovarian injury, including oxidative damage and cell death cascades, resulted in the depletion of follicular reserves and inflammation-related ovarian fibrosis. Cytokine array demonstrated that activated hAECs secreted high levels of paracrine cytokines related to extracellular matrix (ECM) remodeling, angiogenesis, and immunomodulation. An in vivo study showed that the engraftment rate of activated hAECs in damaged ovaries was higher than that of native hAECs. Furthermore, activated hAECs in damaged ovaries had significantly upregulated expression of the antioxidant proteins thioredoxin1/2. In addition, activated hAECs had increased numbers of mature follicles and ameliorated the ovarian microenvironment by promoting angiogenesis and reducing ovarian fibrosis. CONCLUSIONS These results indicated that secondary ovarian damage induced by chemotherapy, including oxidative stress damage, chronic inflammatory response, and ovarian tissue fibrosis should be attended. Prestimulation with the proinflammatory factors TNF-α and IFN-γ could enhance the therapeutic efficacy of hAECs against chemotherapy-induced ovarian dysfunction, which may become a new feasible strategy to improve the therapeutic potential of hAECs in regenerative medicine.
Collapse
Affiliation(s)
- Yating Huang
- The International Peace Maternity and Child Health Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Qiuwan Zhang
- The International Peace Maternity and Child Health Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China.
- Shanghai Key Laboratory of Embryo Original Diseases, 145, Guang-Yuan Road, Shanghai, 200030, China.
| | - Wenjiao Cao
- The International Peace Maternity and Child Health Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Qinyu Zhang
- The International Peace Maternity and Child Health Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Lulu Wang
- The International Peace Maternity and Child Health Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Dongmei Lai
- The International Peace Maternity and Child Health Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China.
- Shanghai Key Laboratory of Embryo Original Diseases, 145, Guang-Yuan Road, Shanghai, 200030, China.
| |
Collapse
|
233
|
Zheng S, Wang H, Han J, Dai X, Lv Y, Sun T, Liu H. Microbiota-derived imidazole propionate inhibits type 2 diabetic skin wound healing by targeting SPNS2-mediated S1P transport. iScience 2023; 26:108092. [PMID: 37876799 PMCID: PMC10590984 DOI: 10.1016/j.isci.2023.108092] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2023] [Revised: 07/20/2023] [Accepted: 09/26/2023] [Indexed: 10/26/2023] Open
Abstract
Imidazole propionate (ImP) is a recently discovered metabolite of T2DM-related gut microbiota. The effect of ImP on T2DM wound healing has not been studied yet. In this research, the changes of ImP-producing bacteria on the skin are firstly evaluated. 16sRNA sequencing results showed that the abundance of ImP-producing bacteria-Streptococcus in the intestine and skin of T2DM mice is significantly increased. Animal experiments show that ImP can inhibit the process of wound healing and inhibit the formation of blood vessels in the process of wound healing. Molecular mechanism research results show that ImP can inhibit S1P secretion mediated by SPNS2, and inhibit the activation of Rho signaling pathway, thereby affecting the angiogenesis process of HUVEC cells. This work also provides a potential drug HMPA that promotes T2DM wound healing.
Collapse
Affiliation(s)
- Shaoting Zheng
- State Key Laboratory of Medicinal Chemical Biology and College of Pharmacy, Nankai University, Tianjin, China
- Tianjin International Joint Academy of Biomedicine, Tianjin, China
| | - Hongqi Wang
- State Key Laboratory of Medicinal Chemical Biology and College of Pharmacy, Nankai University, Tianjin, China
- Tianjin International Joint Academy of Biomedicine, Tianjin, China
| | - Jingxia Han
- State Key Laboratory of Medicinal Chemical Biology and College of Pharmacy, Nankai University, Tianjin, China
| | - Xintong Dai
- State Key Laboratory of Medicinal Chemical Biology and College of Pharmacy, Nankai University, Tianjin, China
| | - Ying Lv
- State Key Laboratory of Medicinal Chemical Biology and College of Pharmacy, Nankai University, Tianjin, China
| | - Tao Sun
- State Key Laboratory of Medicinal Chemical Biology and College of Pharmacy, Nankai University, Tianjin, China
- Tianjin International Joint Academy of Biomedicine, Tianjin, China
| | - Huijuan Liu
- State Key Laboratory of Medicinal Chemical Biology and College of Pharmacy, Nankai University, Tianjin, China
- Tianjin International Joint Academy of Biomedicine, Tianjin, China
| |
Collapse
|
234
|
Youjun D, Huang Y, Lai Y, Ma Z, Wang X, Chen B, Ding X, Tan Q. Mechanisms of resveratrol against diabetic wound by network pharmacology and experimental validation. Ann Med 2023; 55:2280811. [PMID: 37967241 PMCID: PMC10653769 DOI: 10.1080/07853890.2023.2280811] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/27/2023] [Accepted: 10/30/2023] [Indexed: 11/17/2023] Open
Abstract
BACKGROUND Resveratrol (RSV) that possesses anti-oxidative, anti-inflammatory, and pro-angiogenic effects is an effective drug for diabetic wound (DW), while its pharmacological mechanism remains to be elucidated. In this study, we apply network pharmacology and experimental validation approach to reveal the potential mechanism of RSV against DW. METHODS We obtained potential targets for RSV and DW from the publicly available database. Using interaction networks and conducting GO and KEGG pathway enrichment analyses, we constructed target-pathway networks to explore the relationship between RSV and DW. To validate the pharmacological mechanism of RSV, we induced the DW model. RESULTS Ninety overlapped targets between RSV and DW were obtained, and the hub genes of the PPI network included TNF, IL-6, CASP3, MAPK3, VEGFA, IL-1β, AKT1, and JUN. Based on target-pathway networks, the AGE-RAGE signalling pathway was involved in the RSV treatment of DW. Furthermore, in vivo experiments revealed that RSV significantly promoted wound healing in diabetic mice and attenuated the expression of pro-inflammatory cytokines in wound tissue. Meanwhile, RSV could inhibit the AGE-RAGE signalling pathway and thus reduce the activation of NF-κB. CONCLUSION This study initially revealed the biological mechanism of RSV for treating DW through multi-target and multi-pathway. AGE-RAGE, FoxO, MAPK, PI3K-AKT and other signalling pathways may be the main pathways of RSV in treating DW. RSV reduces the inflammatory response by inhibiting the AGE-RAGE signalling pathway, which in turn promotes DW healing.
Collapse
Affiliation(s)
- Ding Youjun
- Nanjing Drum Tower Hospital, Clinical College of Jiangsu University, Nanjing, China
- Department of Emergency Surgery, The Fourth Affiliated Hospital of Jiangsu University (Zhenjiang Fourth People’s Hospital), Zhenjiang, China
| | - Yumeng Huang
- Nanjing Drum Tower Hospital, Clinical College of Jiangsu University, Nanjing, China
| | - Yongxian Lai
- Shanghai Skin Disease Hospital, Tongji University School of Medicine, Shanghai, China
| | - Zhouji Ma
- Nanjing Drum Tower Hospital, Clinical College of Nanjing Medical University, Nanjing, China
| | - Xin Wang
- Jintan Affiliated Hospital of Jiangsu University, Changzhou, China
| | - Bin Chen
- Institute of Plant Resources and Chemistry, Nanjing Research Institute for Comprehensive Utilization of Wild Plants, Nanjing, China
| | - Xiaofeng Ding
- Shanghai Skin Disease Hospital, Tongji University School of Medicine, Shanghai, China
| | - Qian Tan
- Nanjing Drum Tower Hospital, Clinical College of Jiangsu University, Nanjing, China
- Nanjing Drum Tower Hospital, Clinical College of Nanjing Medical University, Nanjing, China
- Nanjing Drum Tower Hospital, The Affiliated Hospital of Nanjing University Medical School, Nanjing, China
| |
Collapse
|
235
|
Fu M, Yang C, Sun G. Recent advances in immunomodulatory hydrogels biomaterials for bone tissue regeneration. Mol Immunol 2023; 163:48-62. [PMID: 37742359 DOI: 10.1016/j.molimm.2023.09.010] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2023] [Revised: 07/27/2023] [Accepted: 09/14/2023] [Indexed: 09/26/2023]
Abstract
There is a high incidence of fractures in clinical practice and therapy. The repairment of critical size defects in the skeletal system remains a huge challenge for surgeons and researchers, which can be overcame by the application of bone tissue-engineered biomaterials. An increasing number of investigations have revealed that the immune system plays a vital role in the repair of bone defects, especially macrophages, which can modulate the integration of biomaterials and bone regeneration in multiple ways. Therefore, it has become increasingly important in regenerative medicine to regulate macrophage polarization to prevent inflammation caused by biomaterial implantation. Recent studies have stressed the importance of hydrogel-based modifications and the incorporation of various cellular and molecular signals for regulating immune responses to promote bone tissue regeneration and integrate biomaterials. In this review, we first elaborate briefly on the described the general physiological mechanism and process of bone tissue regeneration. Then, we summarized the immunomodulatory role macrophages play in bone repair. In addition, the role of hydrogel-based immune modification targeting macrophage modulation in accelerating and enhancing bone tissue regeneration was also discussed. Finally, we highlighted future directions and research strategies related to hydrogel optimization for the regulation of the immune response during bone regeneration and healing.
Collapse
Affiliation(s)
- Mei Fu
- Guixin Sun - Department of Traumatic Surgery, Shanghai East Hospital, School of Medicine, Tongji University, Shanghai 200092, China
| | - Chensong Yang
- Guixin Sun - Department of Traumatic Surgery, Shanghai East Hospital, School of Medicine, Tongji University, Shanghai 200092, China
| | - Guixin Sun
- Guixin Sun - Department of Traumatic Surgery, Shanghai East Hospital, School of Medicine, Tongji University, Shanghai 200092, China.
| |
Collapse
|
236
|
Mao C, Liu X, Guo SW. Reduced endometrial expression of histone deacetylase 3 in women with adenomyosis who complained of heavy menstrual bleeding. Reprod Biomed Online 2023; 47:103288. [PMID: 37690341 DOI: 10.1016/j.rbmo.2023.103288] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2023] [Revised: 07/10/2023] [Accepted: 07/11/2023] [Indexed: 09/12/2023]
Abstract
RESEARCH QUESTION What role, if any, does histone deacetylase 3 (HDAC3) play in adenomyosis-associated heavy menstrual bleeding (HMB)? DESIGN Seventy-two women with adenomyosis-associated HMB were recruited. Of these, 37 women reported moderate/heavy bleeding (MHB) and the remaining 35 women reported excessive bleeding (EXB). The stiffness of adenomyotic lesions and neighbouring endometrial-myometrial interface (EMI) was measured by transvaginal elastosonography, and full-thickness uterine tissue columns were processed for Masson trichrome staining and immunohistochemistry analyses. The protein expression levels of HDAC3 in endometrial cells cultured on substrates of different stiffnesses, and the protein concentrations of nuclear factor-κB (NF-κB) p65 subunit with HDAC3 suppression were evaluated. Mouse experiments were performed to assess the effect of adenomyosis on Hdac3 expression, endometrial repair and bleeding, and to evaluate the effect of HDAC3 inhibition on endometrial repair. RESULTS Compared with controls, the endometrial staining of HDAC3 was significantly lower in women with adenomyosis-associated HMB, concomitant with a greater extent of fibrosis. The stiffness of lesions and neighbouring EMI was significantly higher in the EXB group compared with the MHB group, as was the extent of fibrosis in lesions, their neighboring EMI and endometrium. Expression of HDAC3 was reduced significantly when endometrial epithelial cells were cultured in stiff substrates. Suppression of HDAC3 abrogated the activation and signalling of NF-κB. Mice with induced adenomyosis exhibited reduced Hdac3 staining and elevated fibrosis in endometrium, concomitant with disrupted endometrial repair and more bleeding. Hdac3 inhibition resulted in botched inflammation and increased bleeding. CONCLUSIONS Lesional fibrosis results in reduced endometrial HDAC3 expression and subsequent disruption in NF-κB signalling and inflammation, leading to adenomyosis-associated HMB.
Collapse
Affiliation(s)
- Chenyu Mao
- Department of General Gynaecology, Shanghai Obstetrics and Gynaecology Hospital, Fudan University, Shanghai, China
| | - Xishi Liu
- Department of General Gynaecology, Shanghai Obstetrics and Gynaecology Hospital, Fudan University, Shanghai, China; Shanghai Key Laboratory of Female Reproductive Endocrine-Related Diseases, Fudan University, Shanghai, China
| | - Sun-Wei Guo
- Shanghai Key Laboratory of Female Reproductive Endocrine-Related Diseases, Fudan University, Shanghai, China; Research Institute, Shanghai Obstetrics and Gynaecology Hospital, Fudan University, Shanghai, China.
| |
Collapse
|
237
|
Song L, Chang X, Hu L, Liu L, Wang G, Huang Y, Xu L, Jin B, Song J, Hu L, Zhang T, Wang Y, Xiao Y, Zhang F, Shi M, Liu L, Chen Q, Guo B, Zhou Y. Accelerating Wound Closure With Metrnl in Normal and Diabetic Mouse Skin. Diabetes 2023; 72:1692-1706. [PMID: 37683051 DOI: 10.2337/db23-0173] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/28/2023] [Accepted: 08/21/2023] [Indexed: 09/10/2023]
Abstract
Impaired wound healing and ulcer complications are major causes of morbidity in patients with diabetes. Impaired wound healing is associated with increased inflammation and poor angiogenesis in diabetes patients. Here, we demonstrate that topical administration of a secreted recombinant protein (Meteorin-like [Metrnl]) accelerates wound epithelialization and angiogenesis in mice. We observed a significant increase in Metrnl expression during physiological wound healing; however, its expression remained low during diabetic wound healing. Functionally, the recombinant protein Metrnl significantly accelerated wound closure in normal and diabetic mice models including db/db, high-fat diet/streptozotocin (HFD/STZ), and STZ mice. Mechanistically, keratinocytes secrete quantities of Metrnl to promote angiogenesis; increase endothelial cell proliferation, migration, and tube formation; and enhance macrophage polarization to the M2 type. Meanwhile, M2 macrophages secrete Metrnl to further stimulate angiogenesis. Moreover, the keratinocyte- and macrophage-produced cytokine Metrnl drives postinjury angiogenesis and reepithelialization through activation of AKT phosphorylation (S473) in a KIT receptor tyrosine kinase (c-Kit)-dependent manner. In conclusion, our study suggests that Metrnl has a biological effect in accelerating wound closure through c-Kit-dependent angiogenesis and epithelialization. ARTICLE HIGHLIGHTS
Collapse
Affiliation(s)
- Lingyu Song
- Department of Pathophysiology, Guizhou Medical University, Guiyang, Guizhou, China
- Department of Gastroenterology, Affiliated Hospital of Guizhou Medical University, Guiyang, Guizhou, China
| | - Xuebing Chang
- Department of Pathophysiology, Guizhou Medical University, Guiyang, Guizhou, China
| | - Laying Hu
- Department of Pathophysiology, Guizhou Medical University, Guiyang, Guizhou, China
| | - Lu Liu
- Department of Pathophysiology, Guizhou Medical University, Guiyang, Guizhou, China
| | - Guifang Wang
- Department of Pathophysiology, Guizhou Medical University, Guiyang, Guizhou, China
| | - Yali Huang
- Department of Pathophysiology, Guizhou Medical University, Guiyang, Guizhou, China
| | - Lifen Xu
- Department of Pathology, Affiliated Hospital of Guizhou Medical University, Guiyang, Guizhou, China
| | - Bangming Jin
- Guizhou Province Talent Base of Research on the Pathogenesis and Drug Prevention and Treatment for Common Major Diseases, Guizhou Medical University, Guiyang, Guizhou, China
- Guizhou Provincial Key Laboratory of Pathogenesis and Drug Research on Common Chronic Diseases, Guizhou Medical University, Guiyang, Guizhou, China
| | - Jianying Song
- School of Nursing, Southwest Medical University, Luzhou, Sichuan, China
| | - Lixin Hu
- School of Nursing, Southwest Medical University, Luzhou, Sichuan, China
| | - Tian Zhang
- Department of Pathophysiology, Guizhou Medical University, Guiyang, Guizhou, China
| | - Yuanyuan Wang
- Department of Pathophysiology, Guizhou Medical University, Guiyang, Guizhou, China
| | - Ying Xiao
- Department of Pathophysiology, Guizhou Medical University, Guiyang, Guizhou, China
| | - Fan Zhang
- Department of Pathophysiology, Guizhou Medical University, Guiyang, Guizhou, China
| | - Mingjun Shi
- Department of Pathophysiology, Guizhou Medical University, Guiyang, Guizhou, China
| | - Lingling Liu
- Department of Pathophysiology, Guizhou Medical University, Guiyang, Guizhou, China
| | - Qi Chen
- Department of Endocrinology and Metabolism, The Affiliated Hospital of Southwest Medical University, Luzhou, Sichuan, China
| | - Bing Guo
- Department of Pathophysiology, Guizhou Medical University, Guiyang, Guizhou, China
- Guizhou Province Talent Base of Research on the Pathogenesis and Drug Prevention and Treatment for Common Major Diseases, Guizhou Medical University, Guiyang, Guizhou, China
- Guizhou Provincial Key Laboratory of Pathogenesis and Drug Research on Common Chronic Diseases, Guizhou Medical University, Guiyang, Guizhou, China
| | - Yuxia Zhou
- Department of Pathophysiology, Guizhou Medical University, Guiyang, Guizhou, China
- Guizhou Province Talent Base of Research on the Pathogenesis and Drug Prevention and Treatment for Common Major Diseases, Guizhou Medical University, Guiyang, Guizhou, China
- Guizhou Provincial Key Laboratory of Pathogenesis and Drug Research on Common Chronic Diseases, Guizhou Medical University, Guiyang, Guizhou, China
| |
Collapse
|
238
|
Maduka CV, Alhaj M, Ural E, Habeeb OM, Kuhnert MM, Smith K, Makela AV, Pope H, Chen S, Hix JM, Mallett CL, Chung S, Hakun M, Tundo A, Zinn KR, Hankenson KD, Goodman SB, Narayan R, Contag CH. Polylactide Degradation Activates Immune Cells by Metabolic Reprogramming. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2023; 10:e2304632. [PMID: 37737614 PMCID: PMC10625072 DOI: 10.1002/advs.202304632] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/11/2023] [Revised: 08/04/2023] [Indexed: 09/23/2023]
Abstract
Polylactide (PLA) is the most widely utilized biopolymer in medicine. However, chronic inflammation and excessive fibrosis resulting from its degradation remain significant obstacles to extended clinical use. Immune cell activation has been correlated to the acidity of breakdown products, yet methods to neutralize the pH have not significantly reduced adverse responses. Using a bioenergetic model, delayed cellular changes were observed that are not apparent in the short-term. Amorphous and semi-crystalline PLA degradation products, including monomeric l-lactic acid, mechanistically remodel metabolism in cells leading to a reactive immune microenvironment characterized by elevated proinflammatory cytokines. Selective inhibition of metabolic reprogramming and altered bioenergetics both reduce these undesirable high cytokine levels and stimulate anti-inflammatory signals. The results present a new biocompatibility paradigm by identifying metabolism as a target for immunomodulation to increase tolerance to biomaterials, ensuring safe clinical application of PLA-based implants for soft- and hard-tissue regeneration, and advancing nanomedicine and drug delivery.
Collapse
Affiliation(s)
- Chima V. Maduka
- Comparative Medicine & Integrative BiologyMichigan State UniversityEast LansingMI48824USA
- Department of Biomedical EngineeringMichigan State UniversityEast LansingMI48824USA
- Institute for Quantitative Health Science & EngineeringMichigan State UniversityEast LansingMI48824USA
| | - Mohammed Alhaj
- Department of Chemical Engineering & Materials ScienceMichigan State UniversityEast LansingMI48824USA
| | - Evran Ural
- Department of Biomedical EngineeringMichigan State UniversityEast LansingMI48824USA
- Institute for Quantitative Health Science & EngineeringMichigan State UniversityEast LansingMI48824USA
| | - Oluwatosin M. Habeeb
- Department of Biomedical EngineeringMichigan State UniversityEast LansingMI48824USA
- Institute for Quantitative Health Science & EngineeringMichigan State UniversityEast LansingMI48824USA
| | - Maxwell M. Kuhnert
- Department of Biomedical EngineeringMichigan State UniversityEast LansingMI48824USA
- Institute for Quantitative Health Science & EngineeringMichigan State UniversityEast LansingMI48824USA
| | - Kylie Smith
- Department of Biomedical EngineeringMichigan State UniversityEast LansingMI48824USA
- Institute for Quantitative Health Science & EngineeringMichigan State UniversityEast LansingMI48824USA
| | - Ashley V. Makela
- Department of Biomedical EngineeringMichigan State UniversityEast LansingMI48824USA
- Institute for Quantitative Health Science & EngineeringMichigan State UniversityEast LansingMI48824USA
| | - Hunter Pope
- Department of Biomedical EngineeringMichigan State UniversityEast LansingMI48824USA
- Institute for Quantitative Health Science & EngineeringMichigan State UniversityEast LansingMI48824USA
| | - Shoue Chen
- School of PackagingMichigan State UniversityEast LansingMI48824USA
| | - Jeremy M. Hix
- Institute for Quantitative Health Science & EngineeringMichigan State UniversityEast LansingMI48824USA
| | - Christiane L. Mallett
- Institute for Quantitative Health Science & EngineeringMichigan State UniversityEast LansingMI48824USA
| | - Seock‐Jin Chung
- Department of Biomedical EngineeringMichigan State UniversityEast LansingMI48824USA
- Institute for Quantitative Health Science & EngineeringMichigan State UniversityEast LansingMI48824USA
| | - Maxwell Hakun
- Department of Biomedical EngineeringMichigan State UniversityEast LansingMI48824USA
- Institute for Quantitative Health Science & EngineeringMichigan State UniversityEast LansingMI48824USA
| | - Anthony Tundo
- Department of Biomedical EngineeringMichigan State UniversityEast LansingMI48824USA
- Institute for Quantitative Health Science & EngineeringMichigan State UniversityEast LansingMI48824USA
| | - Kurt R. Zinn
- Department of Biomedical EngineeringMichigan State UniversityEast LansingMI48824USA
- Institute for Quantitative Health Science & EngineeringMichigan State UniversityEast LansingMI48824USA
| | - Kurt D. Hankenson
- Department of Orthopedic SurgeryUniversity of Michigan Medical SchoolAnn ArborMI48109USA
| | - Stuart B. Goodman
- Department of Orthopedic SurgeryStanford UniversityStanfordCA94063USA
- Department of BioengineeringStanford UniversityStanfordCA94305USA
| | - Ramani Narayan
- Department of Chemical Engineering & Materials ScienceMichigan State UniversityEast LansingMI48824USA
| | - Christopher H. Contag
- Department of Biomedical EngineeringMichigan State UniversityEast LansingMI48824USA
- Institute for Quantitative Health Science & EngineeringMichigan State UniversityEast LansingMI48824USA
- Department of Microbiology & Molecular GeneticsMichigan State UniversityEast LansingMI48864USA
| |
Collapse
|
239
|
He T, Bai X, Li Y, Zhang D, Xu Z, Yang X, Hu D, Han J. Insufficient SIRT1 in macrophages promotes oxidative stress and inflammation during scarring. J Mol Med (Berl) 2023; 101:1397-1407. [PMID: 37707556 DOI: 10.1007/s00109-023-02364-x] [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: 02/22/2023] [Revised: 08/18/2023] [Accepted: 08/18/2023] [Indexed: 09/15/2023]
Abstract
Macrophage is a critical regulator in wound healing and scar formation, and SIRT1 is related to macrophage activation and polarization, while the specific mechanism is still unclear. To explore the specific effects of SIRT1 in scarring, we established a skin incision mouse model and LPS-induced inflammation cell model. The expression of SIRT1 in tissue and macrophage was detected, and the level of SIRT1 was changed to observe the downstream effects. LPS-induced macrophages with or without SIRT1 deficiency were used for TMT-based quantitative proteomic analysis. SIRT1 was suppressed in scar while increased in macrophages of scar tissue. And macrophages were proven to be necessary for wound healing. In the early stage of wound healing, knockout of SIRT1 in macrophage could greatly strengthen inflammation and finally promote scarring. NADH-related activities and oxidoreductase activities were differentially expressed in TMT-based quantitative proteomic analysis. We confirmed that ROS production and NOX2 level were elevated after LPS stimulation while the Nrf2 pathway and the downstream proteins, such as Nqo-1 and HO-1, were suppressed. In contrast, the suppression of SIRT1 strengthened this trend. The NF-κB pathway was remarkably activated compared with the control group. Insufficient increase of SIRT1 in macrophage leads to over activated oxidative stress and activates NF-κB pathways, which then promotes inflammation in wound healing and scarring. Further increasing SIRT1 in macrophages could be a promising method to alleviate scarring. KEY MESSAGES: SIRT1 was suppressed in scar while increased in macrophages of scar tissue. Inhibition of SIRT1 in macrophage leads to further activated oxidative stress. SIRT1 is negatively related to oxidative stress in macrophage. The elevation of SIRT1 in macrophage is insufficient during scarring.
Collapse
Affiliation(s)
- Ting He
- Department of Burns and Cutaneous Surgery, Xijing Hospital, Air Force Medical University, 127 Changle West Road, Xi'an, Shaanxi, 710032, China
| | - Xiaozhi Bai
- Department of Burns and Cutaneous Surgery, Xijing Hospital, Air Force Medical University, 127 Changle West Road, Xi'an, Shaanxi, 710032, China
| | - Yan Li
- Department of Burns and Cutaneous Surgery, Xijing Hospital, Air Force Medical University, 127 Changle West Road, Xi'an, Shaanxi, 710032, China
| | - Dongliang Zhang
- Department of Burns and Cutaneous Surgery, Xijing Hospital, Air Force Medical University, 127 Changle West Road, Xi'an, Shaanxi, 710032, China
| | - Zhigang Xu
- Department of Burns and Cutaneous Surgery, Xijing Hospital, Air Force Medical University, 127 Changle West Road, Xi'an, Shaanxi, 710032, China
| | - Xuekang Yang
- Department of Burns and Cutaneous Surgery, Xijing Hospital, Air Force Medical University, 127 Changle West Road, Xi'an, Shaanxi, 710032, China
| | - Dahai Hu
- Department of Burns and Cutaneous Surgery, Xijing Hospital, Air Force Medical University, 127 Changle West Road, Xi'an, Shaanxi, 710032, China.
| | - Juntao Han
- Department of Burns and Cutaneous Surgery, Xijing Hospital, Air Force Medical University, 127 Changle West Road, Xi'an, Shaanxi, 710032, China.
| |
Collapse
|
240
|
Soares GC, Alves APNN, de Sousa AM, Dantas TF, de Barros Silva PG, Júnior EML, de Moraes Filho MO, Paier CRK, Rodrigues FAR, Mota MRL. Evaluation of the healing potential of Nile tilapia skin collagen in traumatic oral ulcers in male rats. Arch Oral Biol 2023; 155:105793. [PMID: 37633029 DOI: 10.1016/j.archoralbio.2023.105793] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2023] [Revised: 08/06/2023] [Accepted: 08/16/2023] [Indexed: 08/28/2023]
Abstract
OBJECTIVE To evaluate the healing potential of Nile tilapia skin collagen using a rat model with experimentally induced traumatic oral ulcers. DESIGN Male Wistar rats were segregated into three experimental groups (n = 8/group/euthanasia day). Ulcers were induced using a dermatological punch on the left buccal mucosa. The rats were then euthanized on days 1, 5, 10, 15, and 20 (ntotal=120 rats). Each group received topical treatment, 2x/day, with 1 % Nile tilapia skin collagen orabase (experimental group), only orabase (negative control), or Oncilom-A® orabase (positive control). Ulcer area, closure percentage, and body mass variation were measured. Slides were prepared for histological analysis, which included Picrosirius red staining (collagen analysis), and immunohistochemistry (platelet endothelial cell adhesion molecule, alpha-smooth muscle actin, and transforming growth factor-beta). RESULTS On day 15, the experimental and positive control groups displayed smaller ulcer areas, a higher percentage of closure, complete re-epithelialization, superior histological repair scores, and a reduced count of polymorphonuclear cells in comparison to the negative control group (p < 0.05). Additionally, the experimental group exhibited an increased number of blood vessels, total collagen (types I and III) and expression of platelet endothelial cell adhesion molecule, alpha-smooth muscle actin, and transforming growth factor-beta relative to the negative and positive control groups (p < 0.05). By day 20, the experimental group showed a more significant weight gain compared to the other groups (p < 0.0001). CONCLUSIONS Nile tilapia skin collagen orabase optimizes the healing of traumatic ulcers by stimulating re-epithelialization, angiogenesis, and collagenesis. Transforming growth factor-beta plays a significant role in this process.
Collapse
Affiliation(s)
- Guilherme Costa Soares
- Department of Dental Clinic, Stomatology and Oral Pathology Sector, Federal University of Ceara, Fortaleza, Brazil
| | | | - Alceu Machado de Sousa
- Department of Dental Clinic, Stomatology and Oral Pathology Sector, Federal University of Ceara, Fortaleza, Brazil
| | - Tales Freitas Dantas
- Department of Dental Clinic, Stomatology and Oral Pathology Sector, Federal University of Ceara, Fortaleza, Brazil
| | - Paulo Goberlânio de Barros Silva
- Department of Dental Clinic, Stomatology and Oral Pathology Sector, Federal University of Ceara, Fortaleza, Brazil; Ceara Cancer Institute, Hospital Haroldo Juaçaba, Fortaleza, Brazil
| | | | - Manoel Odorico de Moraes Filho
- Drug Research and Development Center, Federal University of Ceara, Fortaleza, Brazil; Translational Medicine Postgraduate Program, Federal University of Ceara, Fortaleza, Brazil
| | - Carlos Roberto Koscky Paier
- Drug Research and Development Center, Federal University of Ceara, Fortaleza, Brazil; Translational Medicine Postgraduate Program, Federal University of Ceara, Fortaleza, Brazil
| | - Felipe Augusto Rocha Rodrigues
- Drug Research and Development Center, Federal University of Ceara, Fortaleza, Brazil; Translational Medicine Postgraduate Program, Federal University of Ceara, Fortaleza, Brazil
| | - Mário Rogério Lima Mota
- Department of Dental Clinic, Stomatology and Oral Pathology Sector, Federal University of Ceara, Fortaleza, Brazil.
| |
Collapse
|
241
|
Ben-Chetrit N, Niu X, Sotelo J, Swett AD, Rajasekhar VK, Jiao MS, Stewart CM, Bhardwaj P, Kottapalli S, Ganesan S, Loyher PL, Potenski C, Hannuna A, Brown KA, Iyengar NM, Giri DD, Lowe SW, Healey JH, Geissmann F, Sagi I, Joyce JA, Landau DA. Breast Cancer Macrophage Heterogeneity and Self-renewal are Determined by Spatial Localization. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.10.24.563749. [PMID: 37961223 PMCID: PMC10634790 DOI: 10.1101/2023.10.24.563749] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/15/2023]
Abstract
Tumor-infiltrating macrophages support critical steps in tumor progression, and their accumulation in the tumor microenvironment (TME) is associated with adverse outcomes and therapeutic resistance across human cancers. In the TME, macrophages adopt diverse phenotypic alterations, giving rise to heterogeneous immune activation states and induction of cell cycle. While the transcriptional profiles of these activation states are well-annotated across human cancers, the underlying signals that regulate macrophage heterogeneity and accumulation remain incompletely understood. Here, we leveraged a novel ex vivo organotypic TME (oTME) model of breast cancer, in vivo murine models, and human samples to map the determinants of functional heterogeneity of TME macrophages. We identified a subset of F4/80highSca-1+ self-renewing macrophages maintained by type-I interferon (IFN) signaling and requiring physical contact with cancer-associated fibroblasts. We discovered that the contact-dependent self-renewal of TME macrophages is mediated via Notch4, and its inhibition abrogated tumor growth of breast and ovarian carcinomas in vivo, as well as lung dissemination in a PDX model of triple-negative breast cancer (TNBC). Through spatial multi-omic profiling of protein markers and transcriptomes, we found that the localization of macrophages further dictates functionally distinct but reversible phenotypes, regardless of their ontogeny. Whereas immune-stimulatory macrophages (CD11C+CD86+) populated the tumor epithelial nests, the stroma-associated macrophages (SAMs) were proliferative, immunosuppressive (Sca-1+CD206+PD-L1+), resistant to CSF-1R depletion, and associated with worse patient outcomes. Notably, following cessation of CSF-1R depletion, macrophages rebounded primarily to the SAM phenotype, which was associated with accelerated growth of mammary tumors. Our work reveals the spatial determinants of macrophage heterogeneity in breast cancer and highlights the disruption of macrophage self-renewal as a potential new therapeutic strategy.
Collapse
Affiliation(s)
- Nir Ben-Chetrit
- Sandra and Edward Meyer Cancer Center, Weill Cornell Medicine, New York, NY, USA
- New York Genome Center, New York, NY, USA
- These authors contributed equally
| | - Xiang Niu
- Sandra and Edward Meyer Cancer Center, Weill Cornell Medicine, New York, NY, USA
- New York Genome Center, New York, NY, USA
- These authors contributed equally
- Present address: Genentech, Inc., South San Francisco, CA, USA
| | - Jesus Sotelo
- Sandra and Edward Meyer Cancer Center, Weill Cornell Medicine, New York, NY, USA
- New York Genome Center, New York, NY, USA
| | - Ariel D. Swett
- Sandra and Edward Meyer Cancer Center, Weill Cornell Medicine, New York, NY, USA
- New York Genome Center, New York, NY, USA
| | - Vinagolu K. Rajasekhar
- Orthopedic Service, Department of Surgery, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Maria S. Jiao
- Center of Comparative Medicine and Pathology, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Caitlin M. Stewart
- Sandra and Edward Meyer Cancer Center, Weill Cornell Medicine, New York, NY, USA
- New York Genome Center, New York, NY, USA
| | - Priya Bhardwaj
- Department of Medicine, Weill Cornell Medical College, New York, NY, USA
| | - Sanjay Kottapalli
- Sandra and Edward Meyer Cancer Center, Weill Cornell Medicine, New York, NY, USA
- New York Genome Center, New York, NY, USA
| | - Saravanan Ganesan
- Sandra and Edward Meyer Cancer Center, Weill Cornell Medicine, New York, NY, USA
- New York Genome Center, New York, NY, USA
| | - Pierre-Louis Loyher
- Immunology Program, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Catherine Potenski
- Sandra and Edward Meyer Cancer Center, Weill Cornell Medicine, New York, NY, USA
- New York Genome Center, New York, NY, USA
| | - Assaf Hannuna
- Department of Biological Regulation, Weizmann Institute of Science, Rehovot, Israel
| | - Kristy A. Brown
- Department of Medicine, Weill Cornell Medical College, New York, NY, USA
| | - Neil M. Iyengar
- Department of Medicine, Weill Cornell Medical College, New York, NY, USA
- Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Dilip D. Giri
- Department of Pathology, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Scott W. Lowe
- Cancer Biology and Genetics Program, Memorial Sloan Kettering Cancer Center, New York, NY, USA
- Howard Hughes Medical Institute, Chevy Chase, MD 20815, USA
| | - John H. Healey
- Center of Comparative Medicine and Pathology, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Frederic Geissmann
- Immunology Program, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Irit Sagi
- Department of Biological Regulation, Weizmann Institute of Science, Rehovot, Israel
| | - Johanna A. Joyce
- Department of Oncology and Ludwig Institute for Cancer Research, University of Lausanne, Switzerland
| | - Dan A. Landau
- Sandra and Edward Meyer Cancer Center, Weill Cornell Medicine, New York, NY, USA
- New York Genome Center, New York, NY, USA
| |
Collapse
|
242
|
Mulder PP, Vlig M, Elgersma A, Rozemeijer L, Mastenbroek LS, Middelkoop E, Joosten I, Koenen HJ, Boekema BK. Monocytes and T cells incorporated in full skin equivalents to study innate or adaptive immune reactions after burn injury. Front Immunol 2023; 14:1264716. [PMID: 37901218 PMCID: PMC10611519 DOI: 10.3389/fimmu.2023.1264716] [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: 07/21/2023] [Accepted: 10/03/2023] [Indexed: 10/31/2023] Open
Abstract
Introduction Thermal injury often leads to prolonged and excessive inflammation, which hinders the recovery of patients. There is a notable absence of suitable animal-free models for investigating the inflammatory processes following burn injuries, thereby impeding the development of more effective therapies to improve burn wound healing in patients. Methods In this study, we established a human full skin equivalent (FSE) burn wound model and incorporated human peripheral blood-derived monocytes and T cells. Results Upon infiltration into the FSEs, the monocytes differentiated into macrophages within a span of 7 days. Burn-injured FSEs exhibited macrophages with increased expression of HLA-DR+ and elevated production of IL-8 (CXCL8), in comparison to uninjured FSEs. Among the T cells that actively migrated into the FSEs, the majority were CD4+ and CD25+. These T cells demonstrated augmented expression of markers associated with regulatory T cell, Th1, or Th17 activity, which coincided with significant heightened cytokine production, including IFN-γ, IL-4, IL-6, IL-8, IL-10, IL-12p70, IL-17A, IP-10 (CXCL10), and TGF-β1. Burn injury did not impact the studied effector T cell subsets or cytokine levels. Discussion Collectively, this study represents a significant advancement in the development of an immunocompetent human skin model, specifically tailored for investigating burn-induced innate or adaptive immune reactions at the site of burn injury.
Collapse
Affiliation(s)
- Patrick P.G. Mulder
- Preclinical Research, Association of Dutch Burn Centres (ADBC), Beverwijk, Netherlands
- Laboratory of Medical Immunology, Department of Laboratory Medicine, Radboud University Medical Center, Nijmegen, Netherlands
| | - Marcel Vlig
- Preclinical Research, Association of Dutch Burn Centres (ADBC), Beverwijk, Netherlands
| | - Anouk Elgersma
- Preclinical Research, Association of Dutch Burn Centres (ADBC), Beverwijk, Netherlands
| | - Lotte Rozemeijer
- Preclinical Research, Association of Dutch Burn Centres (ADBC), Beverwijk, Netherlands
| | | | - Esther Middelkoop
- Preclinical Research, Association of Dutch Burn Centres (ADBC), Beverwijk, Netherlands
- Department of Plastic, Reconstructive and Hand Surgery, Amsterdam UMC, VU University Amsterdam, Amsterdam, Netherlands
- Tissue Function and Regeneration, Amsterdam Movement Sciences, Amsterdam, Netherlands
| | - Irma Joosten
- Laboratory of Medical Immunology, Department of Laboratory Medicine, Radboud University Medical Center, Nijmegen, Netherlands
| | - Hans J.P.M. Koenen
- Laboratory of Medical Immunology, Department of Laboratory Medicine, Radboud University Medical Center, Nijmegen, Netherlands
| | - Bouke K.H.L. Boekema
- Preclinical Research, Association of Dutch Burn Centres (ADBC), Beverwijk, Netherlands
- Department of Plastic, Reconstructive and Hand Surgery, Amsterdam UMC, VU University Amsterdam, Amsterdam, Netherlands
| |
Collapse
|
243
|
Huangfu L, Li R, Huang Y, Wang S. The IL-17 family in diseases: from bench to bedside. Signal Transduct Target Ther 2023; 8:402. [PMID: 37816755 PMCID: PMC10564932 DOI: 10.1038/s41392-023-01620-3] [Citation(s) in RCA: 103] [Impact Index Per Article: 51.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2023] [Revised: 06/16/2023] [Accepted: 08/22/2023] [Indexed: 10/12/2023] Open
Abstract
The interleukin-17 (IL-17) family comprises six members (IL-17A-17F), and recently, all of its related receptors have been discovered. IL-17 was first discovered approximately 30 years ago. Members of this family have various biological functions, including driving an inflammatory cascade during infections and autoimmune diseases, as well as boosting protective immunity against various pathogens. IL-17 is a highly versatile proinflammatory cytokine necessary for vital processes including host immune defenses, tissue repair, inflammatory disease pathogenesis, and cancer progression. However, how IL-17 performs these functions remains controversial. The multifunctional properties of IL-17 have attracted research interest, and emerging data have gradually improved our understanding of the IL-17 signaling pathway. However, a comprehensive review is required to understand its role in both host defense functions and pathogenesis in the body. This review can aid researchers in better understanding the mechanisms underlying IL-17's roles in vivo and provide a theoretical basis for future studies aiming to regulate IL-17 expression and function. This review discusses recent progress in understanding the IL-17 signaling pathway and its physiological roles. In addition, we present the mechanism underlying IL-17's role in various pathologies, particularly, in IL-17-induced systemic lupus erythematosus and IL-17-related tumor cell transformation and metastasis. In addition, we have briefly discussed promising developments in the diagnosis and treatment of autoimmune diseases and tumors.
Collapse
Affiliation(s)
- Longjie Huangfu
- School of Stomatology, Harbin Medical University, Harbin, 150001, P. R. China
| | - Ruiying Li
- Department of Oral Pathology, School of Stomatology, Hainan Medical University, Haikou, 571199, P. R. China
| | - Yamei Huang
- Department of Oral Pathology, School of Stomatology, Hainan Medical University, Haikou, 571199, P. R. China
| | - Shan Wang
- Department of Oral Pathology, School of Stomatology, Hainan Medical University, Haikou, 571199, P. R. China.
- Department of Stomatology, The Second Affiliated Hospital of Hainan Medical University, Haikou, 570216, P. R. China.
| |
Collapse
|
244
|
Ou L, Tan X, Qiao S, Wu J, Su Y, Xie W, Jin N, He J, Luo R, Lai X, Liu W, Zhang Y, Zhao F, Liu J, Kang Y, Shao L. Graphene-Based Material-Mediated Immunomodulation in Tissue Engineering and Regeneration: Mechanism and Significance. ACS NANO 2023; 17:18669-18687. [PMID: 37768738 DOI: 10.1021/acsnano.3c03857] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 09/29/2023]
Abstract
Tissue engineering and regenerative medicine hold promise for improving or even restoring the function of damaged organs. Graphene-based materials (GBMs) have become a key player in biomaterials applied to tissue engineering and regenerative medicine. A series of cellular and molecular events, which affect the outcome of tissue regeneration, occur after GBMs are implanted into the body. The immunomodulatory function of GBMs is considered to be a key factor influencing tissue regeneration. This review introduces the applications of GBMs in bone, neural, skin, and cardiovascular tissue engineering, emphasizing that the immunomodulatory functions of GBMs significantly improve tissue regeneration. This review focuses on summarizing and discussing the mechanisms by which GBMs mediate the sequential regulation of the innate immune cell inflammatory response. During the process of tissue healing, multiple immune responses, such as the inflammatory response, foreign body reaction, tissue fibrosis, and biodegradation of GBMs, are interrelated and influential. We discuss the regulation of these immune responses by GBMs, as well as the immune cells and related immunomodulatory mechanisms involved. Finally, we summarize the limitations in the immunomodulatory strategies of GBMs and ideas for optimizing GBM applications in tissue engineering. This review demonstrates the significance and related mechanism of the immunomodulatory function of GBM application in tissue engineering; more importantly, it contributes insights into the design of GBMs to enhance wound healing and tissue regeneration in tissue engineering.
Collapse
Affiliation(s)
- Lingling Ou
- Stomatological Hospital, Southern Medical University, Guangzhou 510280, China
| | - Xiner Tan
- Stomatological Hospital, Southern Medical University, Guangzhou 510280, China
| | - Shijia Qiao
- Stomatological Hospital, Southern Medical University, Guangzhou 510280, China
| | - Junrong Wu
- Stomatological Hospital, Southern Medical University, Guangzhou 510280, China
| | - Yuan Su
- Stomatological Hospital, Southern Medical University, Guangzhou 510280, China
- Stomatology Center, Shunde Hospital, Southern Medical University (The First People's Hospital of Shunde), Foshan 528399, China
| | - Wenqiang Xie
- Stomatological Hospital, Southern Medical University, Guangzhou 510280, China
| | - Nianqiang Jin
- Stomatological Hospital, Southern Medical University, Guangzhou 510280, China
| | - Jiankang He
- Stomatological Hospital, Southern Medical University, Guangzhou 510280, China
| | - Ruhui Luo
- Stomatological Hospital, Southern Medical University, Guangzhou 510280, China
| | - Xuan Lai
- Stomatological Hospital, Southern Medical University, Guangzhou 510280, China
| | - Wenjing Liu
- Stomatological Hospital, Southern Medical University, Guangzhou 510280, China
| | - Yanli Zhang
- Stomatological Hospital, Southern Medical University, Guangzhou 510280, China
| | - Fujian Zhao
- Stomatological Hospital, Southern Medical University, Guangzhou 510280, China
| | - Jia Liu
- Stomatological Hospital, Southern Medical University, Guangzhou 510280, China
| | - Yiyuan Kang
- Stomatological Hospital, Southern Medical University, Guangzhou 510280, China
| | - Longquan Shao
- Stomatological Hospital, Southern Medical University, Guangzhou 510280, China
| |
Collapse
|
245
|
Kamel FO, Karim S, Bafail DAO, Aldawsari HM, Kotta S, Ilyas UK. Hepatoprotective effects of bioactive compounds from traditional herb Tulsi ( Ocimum sanctum Linn) against galactosamine-induced hepatotoxicity in rats. Front Pharmacol 2023; 14:1213052. [PMID: 37860117 PMCID: PMC10582332 DOI: 10.3389/fphar.2023.1213052] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2023] [Accepted: 08/30/2023] [Indexed: 10/21/2023] Open
Abstract
Ocimum sanctum L. (Tulsi; Family: libiaceae), also known as "The Queen of herbs" or "Holy Basil," is an omnipresent, multipurpose plant that has been used in folk medicine of many countries as a remedy against several pathological conditions, including anticancer, antidiabetic, cardio-protective, antispasmodic, diaphoretic, and adaptogenic actions. This study aims to assess O. sanctum L.'s hepatoprotective potential against galactosamine-induced toxicity, as well as investigate bioactive compounds in each extract and identify serum metabolites. The extraction of O. sanctum L as per Ayurveda was simultaneously standardized and quantified for biochemical markers: rutin, ellagic acid, kaempferol, caffeic acid, quercetin, and epicatechin by HPTLC. Hepatotoxicity was induced albino adult rats by intra-peritoneal injection of galactosamine (400 mg/kg). The quantified hydroalcoholic and alcoholic extract of O. sanctum L (100 and 200 mg/kg body weight/day) were compared for evaluation of hepatoprotective potential, which were assessed in terms of reduction in histological damage, change in serum enzymes such as AST, ALT, ALP and increase TBARS. Twenty chemical constituents of serum metabolites of O. sanctum were identified and characterized based on matching recorded mass spectra by GC-MS with those obtained from the library-Wiley/NIST. We evaluated the hepatoprotective activity of various fractions of hydroalcoholic extracts based on the polarity and investigated the activity at each phase (hexane, chloroform, and ethyl acetate) in vitro to determine how they affected the toxicity of CCL4 (40 mM) toward Chang liver cells. The ethyl acetate fraction of the selected plants had a higher hepatoprotective activity than the other fractions, so it was used in vacuum liquid chromatography (VLC). The ethyl acetate fraction contains high amounts of rutin (0.34% w/w), ellagic acid (2.32% w/w), kaempferol (0.017% w/w), caffeic acid (0.005% w/w), quercetin (0.038% w/w), and epicatechin (0.057% w/w) which are responsible for hepatoprotection. In comparison to standard silymarin, isolated bioactive molecules displayed the most significant hepatoprotective activity in Chang liver cells treated to CCl4 toxicity. The significant high hepatoprotection provided by standard silymarin ranged from 77.6% at 100 μg/ml to 83.95% at 200 μg/ml, purified ellagic acid ranged from 70% at 100 μg/ml to 81.33% at 200 μg/ml, purified rutin ranged from 63.4% at 100 μg/ml to 76.34% at 200 μg/ml purified quercetin ranged from 54.33% at 100 μg/ml to 60.64% at 200 μg/ml, purified epicatechin ranged from 53.22% at 100 μg/ml to 65.6% at 200 μg/ml, and purified kaempferol ranged from 52.17% at 100 μg/ml to 60.34% at 200 μg/ml. These findings suggest that the bioactive compounds in O. sanctum L. have significant protective effects against galactosamine-induced hepatotoxicity.
Collapse
Affiliation(s)
- Fatemah O. Kamel
- Department of Clinical Pharmacology, Faculty of Medicine, King Abdulaziz University, Jeddah, Saudi Arabia
| | - Shahid Karim
- Department of Clinical Pharmacology, Faculty of Medicine, King Abdulaziz University, Jeddah, Saudi Arabia
| | - Duaa Abdullah Omer Bafail
- Department of Clinical Pharmacology, Faculty of Medicine, King Abdulaziz University, Jeddah, Saudi Arabia
| | - Hibah Mubarak Aldawsari
- Department of Pharmaceutics, Faculty of Pharmacy, King Abdulaziz University, Jeddah, Saudi Arabia
| | - Sabna Kotta
- Department of Pharmaceutics, Faculty of Pharmacy, King Abdulaziz University, Jeddah, Saudi Arabia
| | - U. K. Ilyas
- Department of Pharmacognosy and Phytochemistry, Moulana College of Pharmacy, Perinthalmanna, Kerala, India
| |
Collapse
|
246
|
Zheng W, Zhou T, Zhang Y, Ding J, Xie J, Wang S, Wang Z, Wang K, Shen L, Zhu Y, Gao C. Simplified α 2-macroglobulin as a TNF-α inhibitor for inflammation alleviation in osteoarthritis and myocardial infarction therapy. Biomaterials 2023; 301:122247. [PMID: 37487780 DOI: 10.1016/j.biomaterials.2023.122247] [Citation(s) in RCA: 15] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2022] [Revised: 06/25/2023] [Accepted: 07/16/2023] [Indexed: 07/26/2023]
Abstract
Tumor necrosis factor α (TNF-α) is a leading proinflammatory cytokine as the master regulator of inflammation in chronic inflammation diseases. Although TNF-α antagonists such as small molecules and peptides are in development, comparable effectiveness in TNF-α neutralization is hardly achieved only with TNF-α capture. In this study, simplified α2-macroglobulin (SM) as a novel TNF-α inhibitor was fabricated to relieve inflammation response by TNF-α capture and internalization with lysosomal degradation. SM was prepared by conjugating a TNF-α-targeting peptide with a receptor binding domain (RBD) derived from α2-macroglobulin through a synthetic biology strategy. SM exhibited effective capture and bioactivity inhibition of TNF-α. Improved endocytosis of TNF-α into lysosomes was observed with SM in macrophages. Even challenged with LPS/IFNγ, the macrophages showed relieved inflammation response with SM treatment. When administrated in chronic inflammation injury in vivo, SM achieved comparable therapeutic efficacy with Infliximab, showing ameliorated cartilage degeneration with relieved inflammation in osteoarthritis (OA) and preserved cardiac function with mitigated myocardium injury in myocardial infarction (MI). These results suggest that SM functioning in TNF-α capture-internalization mechanism might be promising therapeutic alternatives of TNF-α antibodies.
Collapse
Affiliation(s)
- Weiwei Zheng
- MOE Key Laboratory of Macromolecular Synthesis and Functionalization, International Research Center for X Polymers, Department of Polymer Science and Engineering, Zhejiang University, Hangzhou, 310058, China
| | - Tong Zhou
- MOE Key Laboratory of Macromolecular Synthesis and Functionalization, International Research Center for X Polymers, Department of Polymer Science and Engineering, Zhejiang University, Hangzhou, 310058, China
| | - Yuxiang Zhang
- Department of Plastic Surgery, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, 3 East Qingchun Road, Hangzhou, 310016, Zhejiang Province, PR China; Key Laboratory of Tissue Engineering and Regenerative Medicine of Zhejiang Province, Zhejiang University School of Medicine, Hangzhou, 310009, China; Dr. Li Dak Sum Yip Yio Chin Center for Stem Cell and Regenerative Medicine, Zhejiang University, Hangzhou, 310058, China
| | - Jie Ding
- MOE Key Laboratory of Macromolecular Synthesis and Functionalization, International Research Center for X Polymers, Department of Polymer Science and Engineering, Zhejiang University, Hangzhou, 310058, China
| | - Jieqi Xie
- MOE Key Laboratory of Macromolecular Synthesis and Functionalization, International Research Center for X Polymers, Department of Polymer Science and Engineering, Zhejiang University, Hangzhou, 310058, China
| | - Shuqin Wang
- MOE Key Laboratory of Macromolecular Synthesis and Functionalization, International Research Center for X Polymers, Department of Polymer Science and Engineering, Zhejiang University, Hangzhou, 310058, China
| | - Zhaoyi Wang
- MOE Key Laboratory of Macromolecular Synthesis and Functionalization, International Research Center for X Polymers, Department of Polymer Science and Engineering, Zhejiang University, Hangzhou, 310058, China
| | - Kai Wang
- MOE Key Laboratory of Macromolecular Synthesis and Functionalization, International Research Center for X Polymers, Department of Polymer Science and Engineering, Zhejiang University, Hangzhou, 310058, China
| | - Liyin Shen
- MOE Key Laboratory of Macromolecular Synthesis and Functionalization, International Research Center for X Polymers, Department of Polymer Science and Engineering, Zhejiang University, Hangzhou, 310058, China
| | - Yang Zhu
- MOE Key Laboratory of Macromolecular Synthesis and Functionalization, International Research Center for X Polymers, Department of Polymer Science and Engineering, Zhejiang University, Hangzhou, 310058, China
| | - Changyou Gao
- MOE Key Laboratory of Macromolecular Synthesis and Functionalization, International Research Center for X Polymers, Department of Polymer Science and Engineering, Zhejiang University, Hangzhou, 310058, China; Dr. Li Dak Sum Yip Yio Chin Center for Stem Cell and Regenerative Medicine, Zhejiang University, Hangzhou, 310058, China; Center for Healthcare Materials, Shaoxing Institute, Zhejiang University, Shaoxing, 312099, China.
| |
Collapse
|
247
|
Yang L, Bhujel B, Hou Y, Luo J, An SB, Han I, Lee KB. Effective Modulation of Inflammation and Oxidative Stress for Enhanced Regeneration of Intervertebral Discs Using 3D Porous Hybrid Protein Nanoscaffold. ADVANCED MATERIALS (DEERFIELD BEACH, FLA.) 2023; 35:e2303021. [PMID: 37327108 PMCID: PMC10907067 DOI: 10.1002/adma.202303021] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/01/2023] [Revised: 06/11/2023] [Indexed: 06/18/2023]
Abstract
Degeneration of fibrocartilaginous tissues is often associated with complex pro-inflammatory factors. These include reactive oxygen species (ROS), cell-free nucleic acids (cf-NAs), and epigenetic changes in immune cells. To effectively control this complex inflammatory signaling, it developed an all-in-one nanoscaffold-based 3D porous hybrid protein (3D-PHP) self-therapeutic strategy for treating intervertebral disc (IVD) degeneration. The 3D-PHP nanoscaffold is synthesized by introducing a novel nanomaterial-templated protein assembly (NTPA) strategy. 3D-PHP nanoscaffolds that avoid covalent modification of proteins demonstrate inflammatory stimuli-responsive drug release, disc-mimetic stiffness, and excellent biodegradability. Enzyme-like 2D nanosheets incorporated into nanoscaffolds further enabled robust scavenging of ROS and cf-NAs, reducing inflammation and enhancing the survival of disc cells under inflammatory stress in vitro. Implantation of 3D-PHP nanoscaffolds loaded with bromodomain extraterminal inhibitor (BETi) into a rat nucleotomy disc injury model effectively suppressed inflammation in vivo, thus promoting restoration of the extracellular matrix (ECM). The resulting regeneration of disc tissue facilitated long-term pain reduction. Therefore, self-therapeutic and epigenetic modulator-encapsulated hybrid protein nanoscaffold shows great promise as a novel approach to restore dysregulated inflammatory signaling and treat degenerative fibrocartilaginous diseases, including disc injuries, providing hope and relief to patients worldwide.
Collapse
Affiliation(s)
- Letao Yang
- Shanghai Tongji Hospital, School of Life Science and Technologies, Tongji University, Shanghai, 200065, China
- Department of Chemistry and Chemical Biology, Rutgers, The State University of New Jersey, Piscataway, NJ, 08854, USA
| | - Basanta Bhujel
- Department of Neurosurgery, CHA University School of Medicine, CHA Bundang Medical Center, 59 Yaptap-ro, Bundang-gu, Seongnam-si, Gyeonggi-do, 13496, Republic of Korea
| | - Yannan Hou
- Department of Chemistry and Chemical Biology, Rutgers, The State University of New Jersey, Piscataway, NJ, 08854, USA
| | - Jeffrey Luo
- Department of Chemistry and Chemical Biology, Rutgers, The State University of New Jersey, Piscataway, NJ, 08854, USA
| | - Seong Bae An
- Department of Neurosurgery, CHA University School of Medicine, CHA Bundang Medical Center, 59 Yaptap-ro, Bundang-gu, Seongnam-si, Gyeonggi-do, 13496, Republic of Korea
| | - Inbo Han
- Department of Neurosurgery, CHA University School of Medicine, CHA Bundang Medical Center, 59 Yaptap-ro, Bundang-gu, Seongnam-si, Gyeonggi-do, 13496, Republic of Korea
| | - Ki-Bum Lee
- Department of Chemistry and Chemical Biology, Rutgers, The State University of New Jersey, Piscataway, NJ, 08854, USA
| |
Collapse
|
248
|
Manchanda M, Torres M, Inuossa F, Bansal R, Kumar R, Hunt M, Wheelock CE, Bachar-Wikstrom E, Wikstrom JD. Metabolic Reprogramming and Reliance in Human Skin Wound Healing. J Invest Dermatol 2023; 143:2039-2051.e10. [PMID: 37061123 DOI: 10.1016/j.jid.2023.02.039] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2022] [Revised: 01/18/2023] [Accepted: 02/01/2023] [Indexed: 04/17/2023]
Abstract
Impaired skin wound healing is a significant global health issue, especially among the elderly. Wound healing is a well-orchestrated process involving the sequential phases of inflammation, proliferation, and tissue remodeling. Although wound healing is a highly dynamic and energy-requiring process, the role of metabolism remains largely unexplored. By combining transcriptomics and metabolomics of human skin biopsy samples, we mapped the core bioenergetic and metabolic changes in normal acute as well as chronic wounds in elderly subjects. We found upregulation of glycolysis, the tricarboxylic acid cycle, glutaminolysis, and β-oxidation in the later stages of acute wound healing and in chronic wounds. To ascertain the role of these metabolic pathways on wound healing, we targeted each pathway in a wound healing assay as well as in a human skin explant model using metabolic inhibitors and stimulants. Enhancement or inhibition of glycolysis and, to a lesser extent, glutaminolysis had a far greater impact on wound healing than similar manipulations of oxidative phosphorylation and fatty acid β-oxidation. These findings increase the understanding of wound metabolism and identify glycolysis and glutaminolysis as potential targets for therapeutic intervention.
Collapse
Affiliation(s)
- Mansi Manchanda
- Dermatology and Venereology Division, Department of Medicine (Solna), Karolinska Institutet, Stockholm, Sweden
| | - Monica Torres
- Dermatology and Venereology Division, Department of Medicine (Solna), Karolinska Institutet, Stockholm, Sweden; Dermato-Venereology Clinic, Karolinska University Hospital, Stockholm, Sweden
| | - Farydah Inuossa
- Dermatology and Venereology Division, Department of Medicine (Solna), Karolinska Institutet, Stockholm, Sweden
| | - Ritu Bansal
- Dermatology and Venereology Division, Department of Medicine (Solna), Karolinska Institutet, Stockholm, Sweden
| | - Rahul Kumar
- Department of Biotechnology, Indian Institute of Technology Hyderabad, Kandi, India
| | - Matthew Hunt
- Dermatology and Venereology Division, Department of Medicine (Solna), Karolinska Institutet, Stockholm, Sweden
| | - Craig E Wheelock
- Research Division of Physiological Chemistry II, Department of Medical Biochemistry and Biophysics, Karolinska Institutet, Stockholm, Sweden; Department of Respiratory Medicine and Allergy, Karolinska University Hospital, Stockholm, Sweden; Gunma University Initiative for Advanced Research (GIAR), Gunma University, Maebashi, Japan
| | - Etty Bachar-Wikstrom
- Dermatology and Venereology Division, Department of Medicine (Solna), Karolinska Institutet, Stockholm, Sweden
| | - Jakob D Wikstrom
- Dermatology and Venereology Division, Department of Medicine (Solna), Karolinska Institutet, Stockholm, Sweden; Dermato-Venereology Clinic, Karolinska University Hospital, Stockholm, Sweden.
| |
Collapse
|
249
|
Yu J, Cao X, Zhou R, Chen Q, Wang Y. Abnormal brain glucose metabolism patterns in patients with advanced non-small-cell lung cancer after chemotherapy:A retrospective PET study. Brain Res Bull 2023; 202:110751. [PMID: 37625525 DOI: 10.1016/j.brainresbull.2023.110751] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2023] [Revised: 07/24/2023] [Accepted: 08/23/2023] [Indexed: 08/27/2023]
Abstract
PURPOSE This study was designed to investigate the acute or chronic post-chemotherapy effect and different chemotherapy cycles effect on brain glucose metabolism. METHODS A total of seventy-three patients who received chemotherapy after being diagnosed with advanced non-small-cell lung cancer (NSCLC) and underwent 18F-Fluorodeoxyglucose positron emission tomography/computed tomography (18F-FDG PET/CT) scan at Nuclear Medicine Department of the Fifth Hospital of Sun Yat-sen University between September 2017 and August 2022 were included. Seventy-two healthy control patients who underwent whole-body 18F-FDG PET/CT scans at our department, without any evidence of malignancy and confirmed by follow-up visits, were included. Advanced NSCLC patients were classified into six arms: short-to-long course (chemotherapy cycles under 4, between 5 and 8 and more than 8) in acute chemotherapy effect (AC) group (scanned 18F-FDG PET/CT within 6 months post-chemotherapy) or chronic chemotherapy effect (CC) group (the interval between scanning and the last chemotherapy session more than six months). Statistical Parametric Mapping (SPM) analysis between patients' groups and healthy controls' brain 18F-FDG PET was performed (uncorrected p ˂ 0.001 with cluster size above 20 contiguous voxels). RESULTS There were no significant differences between patients' groups and healthy controls in age, gender and body mass index (BMI). SPM PET analyses revealed anomalous brain metabolic activity in different groups (p ˂ 0.001). Short-course + AC group exhibited hypermetabolism in the cerebellum and widespread hypometabolism in bilateral frontal lobe predominantly. Only hypometabolic brain regions were observed in middle-course + AC patients. Long-course + AC group displayed a greater number of abnormalities. Notably, these metabolic abnormalities tended to decrease in CC groups versus AC groups across all courses. CONCLUSION Our study revealed that patients with advanced NSCLC who underwent chemotherapy exhibited persistent abnormal brain metabolism patterns during continuous chemotherapy and these abnormalities tended to recover after completion of chemotherapy over time, but without correlation to an increasing number of chemotherapy cycles. 18F-FDG PET/CT may serve as a possible modality for evaluating brain function and guiding appropriate treatment timing.
Collapse
Affiliation(s)
- Jie Yu
- Department of Nuclear Medicine, The Fifth Affiliated Hospital of Sun Yat-sen University, Sun Yat-sen University, Zhuhai, Guangdong, China
| | - Xiaoling Cao
- Department of Nuclear Medicine, The Fifth Affiliated Hospital of Sun Yat-sen University, Sun Yat-sen University, Zhuhai, Guangdong, China
| | - Renwei Zhou
- Department of Nuclear Medicine, The Fifth Affiliated Hospital of Sun Yat-sen University, Sun Yat-sen University, Zhuhai, Guangdong, China
| | - Qingling Chen
- Department of Nuclear Medicine, The Fifth Affiliated Hospital of Sun Yat-sen University, Sun Yat-sen University, Zhuhai, Guangdong, China
| | - Ying Wang
- Department of Nuclear Medicine, The Fifth Affiliated Hospital of Sun Yat-sen University, Sun Yat-sen University, Zhuhai, Guangdong, China.
| |
Collapse
|
250
|
Ren LL, Miao H, Wang YN, Liu F, Li P, Zhao YY. TGF-β as A Master Regulator of Aging-Associated Tissue Fibrosis. Aging Dis 2023; 14:1633-1650. [PMID: 37196129 PMCID: PMC10529747 DOI: 10.14336/ad.2023.0222] [Citation(s) in RCA: 33] [Impact Index Per Article: 16.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2023] [Accepted: 02/22/2023] [Indexed: 05/19/2023] Open
Abstract
Fibrosis is the abnormal accumulation of extracellular matrix proteins such as collagen and fibronectin. Aging, injury, infections, and inflammation can cause different types of tissue fibrosis. Numerous clinical investigations have shown a correlation between the degree of liver and pulmonary fibrosis in patients and telomere length and mitochondrial DNA content, both of which are signs of aging. Aging involves the gradual loss of tissue function over time, which results in the loss of homeostasis and, ultimately, an organism's fitness. A major feature of aging is the accumulation of senescent cells. Senescent cells abnormally and continuously accumulate in the late stages of life, contributing to age-related fibrosis and tissue deterioration, among other aging characteristics. Furthermore, aging generates chronic inflammation, which results in fibrosis and decreases organ function. This finding suggests that fibrosis and aging are closely related. The transforming growth factor-beta (TGF-β) superfamily plays a crucial role in the physiological and pathological processes of aging, immune regulation, atherosclerosis, and tissue fibrosis. In this review, the functions of TGF-β in normal organs, aging, and fibrotic tissues is discussed: TGF-β signalling is altered with age and is an indicator of pathology associated with tissue fibrosis. In addition, this review discusses the potential targeting of noncoding.
Collapse
Affiliation(s)
- Li-Li Ren
- School of Pharmacy, Zhejiang Chinese Medical University, Hangzhou, Zhejiang, China.
| | - Hua Miao
- School of Pharmacy, Zhejiang Chinese Medical University, Hangzhou, Zhejiang, China.
| | - Yan-Ni Wang
- School of Pharmacy, Zhejiang Chinese Medical University, Hangzhou, Zhejiang, China.
| | - Fei Liu
- Department of Urology, Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China.
| | - Ping Li
- Beijing Key Lab for Immune-Mediated Inflammatory Diseases, Institute of Clinical Medical Science, Department of Nephrology, China-Japan Friendship Hospital, Beijing, China.
| | - Ying-Yong Zhao
- School of Pharmacy, Zhejiang Chinese Medical University, Hangzhou, Zhejiang, China.
| |
Collapse
|