1
|
Balakrishnan R, Subbarayan R, Shrestha R, Chauhan A, Krishnamoorthy L. Exploring platelet-derived microvesicles in vascular regeneration: unraveling the intricate mechanisms and molecular mediators. Mol Biol Rep 2024; 51:393. [PMID: 38446325 DOI: 10.1007/s11033-024-09302-1] [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: 12/22/2023] [Accepted: 01/30/2024] [Indexed: 03/07/2024]
Abstract
Microvesicles (MVs) serve as biomarkers and transmitters for cell communication and also act as essential contributors to diseases. Platelets release microvesicles when activated voluntarily, making them a significant source. Platelet-derived microvesicles possess a range of characteristics similar to their parent cells and were shown to exert regulatory impacts on vascular and immunological cells. MVs can alter the activity of recipient cells by transferring their internal components. Furthermore, it has been identified that microvesicles derived from platelets possess the ability to exert immunomodulatory effects on different kinds of cells. Recent research has shown that microvesicles have a bidirectional influence of harming and preventing the receptor cells. Nevertheless, the specific characteristics of the active molecules responsible for this phenomenon are still unknown. The primary focus of this review was to explore the mechanism of vascular tissue regeneration and the specific molecules that play a role in mediating various biological effects throughout this process. These molecules exert their effects by influencing autophagy, apoptosis, and inflammatory pathways.
Collapse
Affiliation(s)
- Ranjith Balakrishnan
- Centre for Advanced Biotherapeutics and Regenerative Medicine, FAHS, Chettinad Hospital and Research Institute, Chettinad Academy of Research and Education, Kelambakkam, India
| | - Rajasekaran Subbarayan
- Centre for Advanced Biotherapeutics and Regenerative Medicine, FAHS, Chettinad Hospital and Research Institute, Chettinad Academy of Research and Education, Kelambakkam, India.
| | | | - Ankush Chauhan
- Faculty of Allied Health Sciences, Centre for Herbal Pharmacology and Environmental Sustainability, Chettinad Hospital and Research Institute, Chettinad Academy of Research and Education, Kelambakkam, India
| | - Loganathan Krishnamoorthy
- FAHS, Chettinad Hospital and Research Institute, Chettinad Academy of Research and Education, Kelambakkam, India
| |
Collapse
|
2
|
Zhang Y, Fu X, Wang L, Guo X, Dong B. Sorption of phenols and flavonoids on activated charcoal improves protein metabolism, antioxidant status, immunity, and intestinal morphology in broilers. Front Vet Sci 2024; 10:1327455. [PMID: 38274659 PMCID: PMC10808307 DOI: 10.3389/fvets.2023.1327455] [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: 10/25/2023] [Accepted: 12/20/2023] [Indexed: 01/27/2024] Open
Abstract
Previous studies have revealed that activated charcoal sorption of Chinese herbal extracts is more effective than activated charcoal. The present study was designed to investigate whether phenols and flavonoids have an effect on nutrient metabolism, antioxidant activity, immunity, and intestinal morphology in broilers. Seven diets [basal diet (CON); CON supplemented with 450 mg/kg of activated charcoal (AC); CON supplemented with 250, 500, 750, 1,000, or 7,500 mg/kg of phenolic acids and flavonoids (PF) to AC (PFAC)]. PFAC was the complex of AC sorption of PF in the ratio of 9:1. These dietary treatments for broilers lasted for 42 days. Results showed that at d 21, all doses of PFAC altered serum levels of total protein, albumin, and creatinine compared to AC (p < 0.05). Both PFAC and AC altered HDL-, LDL-, and VLDL-cholesterol levels compared to CON (p < 0.05). PFAC at 500 mg/kg (450 mg/kg AC+ 50 mg/kg phenolic acids and flavonoids) increased serum IgA and IgM (p < 0.05), but AC at 450 mg/kg did not, compared to CON. At d 42, breast and thigh muscles of PFAC-treated broilers had higher free radical scavenging activities compared to CON (p < 0.05), but AC had no such effect. PFAC at 500 mg/kg increased villus height in the duodenum, jejunum, and ileum compared to CON (p < 0.05), but AC had no such response. PFAC at 500 mg/kg effectively improved protein and lipid metabolism, antioxidant status, and intestinal morphology, but AC had no such effect at a similar dose. Excessive PFAC (7,500 mg/kg) showed no significant side effects on broiler growth, liver damage, or hematology. These results suggest that phenols and flavonoids, in cooperation with activated charcoal, provide the majority of the functions of the herbal extract from multiple Chinese medicinal herbs.
Collapse
Affiliation(s)
- Ying Zhang
- State Key Laboratory of Animal Nutrition, College of Animal Science, China Agricultural University, Beijing, China
- Institute for Hepatology National Clinical Research Center for Infectious Disease, Shenzhen Third People’s Hospital, Shenzhen, China
| | - Xiaoqi Fu
- Institute for Hepatology National Clinical Research Center for Infectious Disease, Shenzhen Third People’s Hospital, Shenzhen, China
- Plateau Animal Nutrition and Feed Laboratory, Tibet Agriculture and Animal Husbandry University, Nyingchi, China
| | - Lixue Wang
- State Key Laboratory of Animal Nutrition, College of Animal Science, China Agricultural University, Beijing, China
| | - Xiangyue Guo
- State Key Laboratory of Animal Nutrition, College of Animal Science, China Agricultural University, Beijing, China
| | - Bing Dong
- State Key Laboratory of Animal Nutrition, College of Animal Science, China Agricultural University, Beijing, China
| |
Collapse
|
3
|
Dutta SD, Ganguly K, Patil TV, Randhawa A, Lim KT. Unraveling the potential of 3D bioprinted immunomodulatory materials for regulating macrophage polarization: State-of-the-art in bone and associated tissue regeneration. Bioact Mater 2023; 28:284-310. [PMID: 37303852 PMCID: PMC10248805 DOI: 10.1016/j.bioactmat.2023.05.014] [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: 11/17/2022] [Revised: 04/29/2023] [Accepted: 05/20/2023] [Indexed: 06/13/2023] Open
Abstract
Macrophage-assisted immunomodulation is an alternative strategy in tissue engineering, wherein the interplay between pro-inflammatory and anti-inflammatory macrophage cells and body cells determines the fate of healing or inflammation. Although several reports have demonstrated that tissue regeneration depends on spatial and temporal regulation of the biophysical or biochemical microenvironment of the biomaterial, the underlying molecular mechanism behind immunomodulation is still under consideration for developing immunomodulatory scaffolds. Currently, most fabricated immunomodulatory platforms reported in the literature show regenerative capabilities of a particular tissue, for example, endogenous tissue (e.g., bone, muscle, heart, kidney, and lungs) or exogenous tissue (e.g., skin and eye). In this review, we briefly introduced the necessity of the 3D immunomodulatory scaffolds and nanomaterials, focusing on material properties and their interaction with macrophages for general readers. This review also provides a comprehensive summary of macrophage origin and taxonomy, their diverse functions, and various signal transduction pathways during biomaterial-macrophage interaction, which is particularly helpful for material scientists and clinicians for developing next-generation immunomodulatory scaffolds. From a clinical standpoint, we briefly discussed the role of 3D biomaterial scaffolds and/or nanomaterial composites for macrophage-assisted tissue engineering with a special focus on bone and associated tissues. Finally, a summary with expert opinion is presented to address the challenges and future necessity of 3D bioprinted immunomodulatory materials for tissue engineering.
Collapse
Affiliation(s)
- Sayan Deb Dutta
- Department of Biosystems Engineering, Kangwon National University, Chuncheon, 24341, Republic of Korea
- Institute of Forest Science, Kangwon National University, Chuncheon, 24341, Republic of Korea
| | - Keya Ganguly
- Department of Biosystems Engineering, Kangwon National University, Chuncheon, 24341, Republic of Korea
| | - Tejal V. Patil
- Department of Biosystems Engineering, Kangwon National University, Chuncheon, 24341, Republic of Korea
- Interdisciplinary Program in Smart Agriculture, Kangwon National University, Chuncheon, 24341, Republic of Korea
| | - Aayushi Randhawa
- Department of Biosystems Engineering, Kangwon National University, Chuncheon, 24341, Republic of Korea
- Interdisciplinary Program in Smart Agriculture, Kangwon National University, Chuncheon, 24341, Republic of Korea
| | - Ki-Taek Lim
- Department of Biosystems Engineering, Kangwon National University, Chuncheon, 24341, Republic of Korea
- Institute of Forest Science, Kangwon National University, Chuncheon, 24341, Republic of Korea
- Interdisciplinary Program in Smart Agriculture, Kangwon National University, Chuncheon, 24341, Republic of Korea
| |
Collapse
|
4
|
Lu YN, Wang L, Zhang YZ. The promising roles of macrophages in geriatric hip fracture. Front Cell Dev Biol 2022; 10:962990. [PMID: 36092716 PMCID: PMC9458961 DOI: 10.3389/fcell.2022.962990] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2022] [Accepted: 07/26/2022] [Indexed: 11/13/2022] Open
Abstract
As aging becomes a global burden, the incidence of hip fracture (HF), which is the most common fracture in the elderly population and can be fatal, is rapidly increasing, and its extremely high fatality rate places significant medical and financial burdens on patients. Fractures trigger a complex set of immune responses, and recent studies have shown that with aging, the immune system shows decreased activity or malfunctions in a process known as immune senescence, leading to disease and death. These phenomena are the reasons why elderly individuals typically exhibit chronically low levels of inflammation and increased rates of infection and chronic disease. Macrophages, which are key players in the inflammatory response, are critical in initiating the inflammatory response, clearing pathogens, controlling the innate and adaptive immune responses and repairing damaged tissues. Tissue-resident macrophages (TRMs) are widely present in tissues and perform immune sentinel and homeostatic functions. TRMs are combinations of macrophages with different functions and phenotypes that can be directly influenced by neighboring cells and the microenvironment. They form a critical component of the first line of defense in all tissues of the body. Immune system disorders caused by aging could affect the biology of macrophages and thus the cascaded immune response after fracture in various ways. In this review, we outline recent studies and discuss the potential link between monocytes and macrophages and their potential roles in HF in elderly individuals.
Collapse
Affiliation(s)
- Yi-ning Lu
- Department of Orthopedic Research Center, The Third Hospital of Hebei Medical University, Shijiazhuang, China
- Department of Orthopedic Surgery, The Third Hospital of Hebei Medical University, Shijiazhuang, China
| | - Ling Wang
- Department of Orthopedic Research Center, The Third Hospital of Hebei Medical University, Shijiazhuang, China
- Department of Orthopedic Oncology, The Third Hospital of Hebei Medical University, Shijiazhuang, China
- *Correspondence: Ying-ze Zhang, ; Ling Wang,
| | - Ying-ze Zhang
- Department of Orthopedic Research Center, The Third Hospital of Hebei Medical University, Shijiazhuang, China
- Department of Orthopedic Surgery, The Third Hospital of Hebei Medical University, Shijiazhuang, China
- *Correspondence: Ying-ze Zhang, ; Ling Wang,
| |
Collapse
|
5
|
Inducible Animal Models of Skin Fibrosis; Updated Review of the Literature. JORJANI BIOMEDICINE JOURNAL 2022. [DOI: 10.52547/jorjanibiomedj.10.2.69] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/17/2023]
|
6
|
Molecular Mechanism of Traditional Chinese Ointment of Xuzhou Qufu Shengji in Infected Wounds. EVIDENCE-BASED COMPLEMENTARY AND ALTERNATIVE MEDICINE 2022; 2022:4116563. [PMID: 35069758 PMCID: PMC8769828 DOI: 10.1155/2022/4116563] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 10/27/2021] [Revised: 11/29/2021] [Accepted: 12/15/2021] [Indexed: 11/19/2022]
Abstract
Background Xuzhou Qufu Shengji Ointment (QFSJO) has been used in hospital and private medication for more than 30 years to treat the infective wounds after trauma. However, molecular investigation is lacking. This study used rats to explore the healing mechanism of QFSJO in promoting wound healing in human. Methods One circular incision was individually generated on the back of 30 rats in three groups and challenged with 108 CFU (0.3 mL) of Staphylococcus aureus. Then, one of the trauma groups was treated with QFSJO gauze, and the control group was covered with a piece of Vaseline gauze, while the western medicine group was treated with erythromycin in a similar way. The dressing change of all the groups was performed once a day for three weeks. The anti-inflammation and proangiogenesis of QFSJO were evaluated by enzyme-linked immunosorbent assay (ELISA). The levels of angiogenesis associated factors, vascular endothelial growth factor (VEGF) and basic fibroblast growth factor (b-FGF), hydroxyproline, and hemoglobin, were measured according to ELISA. The immunohistochemistry of CD31 and CD34 expression in granulation tissue was demonstrated and quantitatively analyzed for angiogenesis in granulation tissue in sites. Results A faster wound healing ratio was observed in QFSJO-dressing-treated group than Vaseline- and erythrocin-treated groups. ELISA results showed that QFSJO promoted VEGF and b-FGF levels significantly in early stage of wound healing. QFSJO dressing group also showed an enhanced hydroxyproline and hemoglobin in granulation tissue. The expressions of CD31 and CD34 in granulation tissue of QFSJO group were higher than in the Vaseline and erythrocin groups. Conclusion QFSJO improved the healing rate of the infective wounds by promoting the angiogenesis of granulation tissue and inhibiting the inflammation of the trauma tissue. Our finding suggests that QFSJO is able to help angiogenic capillary sprouts for collagen accumulates in the granulation tissue.
Collapse
|
7
|
Huang J, Liu X, Hou Y, Liu Y, Liao K, Xie N, Deng K. Macrophage polarisation in caesarean scar diverticulum. J Clin Pathol 2022; 76:379-383. [PMID: 34980638 DOI: 10.1136/jclinpath-2021-207926] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2021] [Accepted: 11/30/2021] [Indexed: 11/04/2022]
Abstract
AIMS To determine immunohistochemical features and correlations between M1/M2 polarisation status with disease severity of post-caesarean scar diverticulum (CSD). METHODS Histological and immunohistological stainings were performed and inflammatory (CD16, CD163 and tumour necrosis factor-α (TNF-α)), fibrosis (α-smooth muscle actin (α-SMA)) and angiogenic (CD31) markers were examined in uterine tissues collected from patients with uterine scar diverticula (CSD) (n=37) and caesarean section (CS) (n=3). RESULTS CSD tissues have higher expression of α-SMA, TNF-α, CD16 and CD31 and lower expression of CD163 than CS tissue (p<0.05). Compared with adjacent tissues, thick-walled blood vessels, glands and fibrotic sites have higher expression of α-SMA, TNF-α and CD16. Statistical correlation was observed between the expression of CD16 and TNF-α (R=0.693, p<0.001), α-SMA (R=0.404, p<0.05) and CD31 (R=0.253, p<0.05) in CSD tissues, especially with the ratio of CD16/CD163 (R=0.590, p<0.01). A more significant difference was observed between the expression of CD16/CD163 and α-SMA (R=0.556, p<0.001), TNF-α (R=0.633, p<0.0001) and CD31 (R=0.336, p<0.05). CONCLUSIONS In this study, TNF-α, α-SMA, CD16 and CD31 proteins were overexpressed in all CSD cases, and CD16/CD163 was positively correlated with tissue inflammation, fibrosis and neovascularisation. Abnormal mononuclear macrophage infiltration may be involved in the origin and progression of CSD.
Collapse
Affiliation(s)
- Jinfa Huang
- Department of Gynecology, Shunde Hospital of Southern Medical University, Foshan, Guangdong, China
| | - Xiaochun Liu
- Department of Gynecology, Southern Medical University Affiliated Maternal & Child Health Hospital of Foshan, Foshan, Guangdong, China
| | - Yi Hou
- Department of Bioinformatics, Guangzhou Regenerative Medicine and Health-Guangdong Laboratory, Guangzhou, Guangdong, China
| | - Yixuan Liu
- Department of Gynecology, Shunde Hospital of Southern Medical University, Foshan, Guangdong, China
| | - Kedan Liao
- Department of Gynecology, Shunde Hospital of Southern Medical University, Foshan, Guangdong, China
| | - Ning Xie
- Department of Gynecology, Shunde Hospital of Southern Medical University, Foshan, Guangdong, China
| | - Kaixian Deng
- Department of Gynecology, Shunde Hospital of Southern Medical University, Foshan, Guangdong, China
| |
Collapse
|
8
|
Zhang XY, Chang HM, Yi Y, Zhu H, Liu RZ, Leung PCK. BMP6 increases CD68 expression by up-regulating CTGF expression in human granulosa-lutein cells. Mol Cell Endocrinol 2021; 536:111414. [PMID: 34314740 DOI: 10.1016/j.mce.2021.111414] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/19/2021] [Revised: 07/22/2021] [Accepted: 07/23/2021] [Indexed: 11/20/2022]
Abstract
Bone morphogenetic protein 6 (BMP6) and connective tissue growth factor (CTGF) are critical growth factors required for normal follicular development and luteal function. Cluster of Differentiation 68 (CD68) is an intraovarian marker of macrophages that plays an important role in modulating the physiological regression of the corpus luteum. The aim of this study was to investigate the effect of BMP6 on the expression of CTGF and the subsequent increase in CD68 expression as well as its underlying mechanisms. Primary and immortalized (SVOG) human granulosa cells obtained from infertile women undergoing in vitro fertilization treatment were used as cell models to conduct the in vitro experiments. Our results showed that BMP6 treatment significantly increased the expression levels of CTGF and CD68. Using BMP type I receptor inhibitors (dorsomorphin, DMH-1 and SB431542), we demonstrated that both activin receptor-like kinase (ALK)2 and ALK3 are involved in BMP6-induced stimulatory effects on the expression of CTGF and CD68. Additionally, SMAD4-knock down reversed the BMP6-induced up-regulation of CTGF and CD68, indicating that the canonical SMAD signaling pathway is required for these effects. Moreover, CTGF-knock down abolished the BMP6-induced up-regulation of CD68 expression. These findings indicate that intrafollicular CTGF mediates BMP6-induced increases in CD68 expression through the ALK2/ALK3-mediated SMAD-dependent signaling pathway.
Collapse
Affiliation(s)
- Xin-Yue Zhang
- Center for Reproductive Medicine, The First Hospital of Jilin University, Changchun, Jilin, China; Department of Obstetrics and Gynaecology, BC Children's Hospital Research Institute, University of British Columbia, Vancouver, British Columbia, V5Z 4H4, Canada
| | - Hsun-Ming Chang
- Department of Obstetrics and Gynaecology, BC Children's Hospital Research Institute, University of British Columbia, Vancouver, British Columbia, V5Z 4H4, Canada
| | - Yuyin Yi
- Department of Obstetrics and Gynaecology, BC Children's Hospital Research Institute, University of British Columbia, Vancouver, British Columbia, V5Z 4H4, Canada
| | - Hua Zhu
- Department of Obstetrics and Gynaecology, BC Children's Hospital Research Institute, University of British Columbia, Vancouver, British Columbia, V5Z 4H4, Canada
| | - Rui-Zhi Liu
- Center for Reproductive Medicine, The First Hospital of Jilin University, Changchun, Jilin, China.
| | - Peter C K Leung
- Department of Obstetrics and Gynaecology, BC Children's Hospital Research Institute, University of British Columbia, Vancouver, British Columbia, V5Z 4H4, Canada.
| |
Collapse
|
9
|
Yu Y, Wu H, Zhang Q, Ogawa R, Fu S. Emerging insights into the immunological aspects of keloids. J Dermatol 2021; 48:1817-1826. [PMID: 34549462 DOI: 10.1111/1346-8138.16149] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2021] [Revised: 08/18/2021] [Accepted: 08/25/2021] [Indexed: 12/21/2022]
Abstract
A special kind of scar, keloid, sometimes grows huge, disturbing patients in different ways. We discussed the pathogenesis of keloids and found researches about fibroblasts and collagen disorders, with little emphasis on immunity. Coupled with few effective treatments in keloid at present, we have focused on the immunological mechanisms of keloids with an aim to unravel some new therapeutic approaches in the future. In this review, the immunological processes are separately illustrated by the classification of different immune cells. In addition, we also discuss possible reasons for the repeated recurrence of keloids, the phenomenon of cell talks, and inflammation-related signal pathways involved in the pathogenesis of keloids.
Collapse
Affiliation(s)
- Yangyiyi Yu
- Hunan Key Laboratory of Medical Epigenomics, Department of Dermatology, Second Xiangya Hospital, Central South University, Changsha, China
| | - Haijing Wu
- Hunan Key Laboratory of Medical Epigenomics, Department of Dermatology, Second Xiangya Hospital, Central South University, Changsha, China
| | - Qing Zhang
- Hunan Key Laboratory of Medical Epigenomics, Department of Dermatology, Second Xiangya Hospital, Central South University, Changsha, China
| | - Rei Ogawa
- Department of Plastic, Reconstructive and Aesthetic Surgery, Nippon Medical School, Tokyo, Japan
| | - Siqi Fu
- Hunan Key Laboratory of Medical Epigenomics, Department of Dermatology, Second Xiangya Hospital, Central South University, Changsha, China
| |
Collapse
|
10
|
Zhang SM, Wei CY, Wang Q, Wang L, Lu L, Qi FZ. M2-polarized macrophages mediate wound healing by regulating connective tissue growth factor via AKT, ERK1/2, and STAT3 signaling pathways. Mol Biol Rep 2021; 48:6443-6456. [PMID: 34398425 DOI: 10.1007/s11033-021-06646-w] [Citation(s) in RCA: 19] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2021] [Accepted: 08/11/2021] [Indexed: 02/07/2023]
Abstract
BACKGROUND Timely and sufficient M1 recruitment and M2 polarization are necessary for fibrosis during wound healing. The mechanism of how M2 mediates wound healing is worth exploring. Abnormally up-regulated connective tissue growth factor (CTGF) influences multiple organ fibrosis, including cardiac, pulmonary, hepatic, renal, and cutaneous fibrosis. Previous studies reported that M2 contributed to hepatic and renal fibrosis by secreting CTGF. It is worth discussing if M2 regulates fibrosis through secreting CTGF in wound healing. METHODS AND RESULTS We established the murine wound model and inhibited macrophages during proliferation phase with clodronate liposomes in vivo. Macrophages depletion led to down-regulation of wound healing rates, collagen deposition, as well as expression of collagen 1/3 and Ki67. M2 was induced by interleukin-4 (IL-4) and measured by flow cytometry in vitro. Secreted pro-fibrotic and anti-fibrotic factors were tested by enzyme-linked immunosorbent assay (ELISA). M2 was polarized, which producing more CTGF, transforming growth factor-beta1 (TGF-β1), and IL-6, as well as less tumor necrosis factor-α (TNF-α) and IL-10. M2 CTGF gene was blocked using siCTGF. Effects of M2 on fibroblasts activities were detected by cell counting kit 8 (CCK8) and cellular wound healing assay. Expressions of related signaling pathway were assessed by western blotting. Blockade of CTGF in M2 deactivated fibroblasts proliferation and migration by regulating AKT, ERK1/2, and STAT3 pathway. Recombinant CTGF restored these effects. CONCLUSIONS Our research, for the first time, indicated that M2 promoted wound healing by secreting CTGF, which further mediating proliferation and migration of fibroblasts via AKT, ERK1/2, and STAT3 pathway.
Collapse
Affiliation(s)
- Si-Min Zhang
- Department of Plastic and Reconstructive Surgery, Zhongshan Hospital Fudan University, 180 Fenglin Road, Xuhui District, Shanghai, 200032, People's Republic of China
| | - Chuan-Yuan Wei
- Department of Plastic and Reconstructive Surgery, Zhongshan Hospital Fudan University, 180 Fenglin Road, Xuhui District, Shanghai, 200032, People's Republic of China
| | - Qiang Wang
- Department of Plastic and Reconstructive Surgery, Zhongshan Hospital Fudan University, 180 Fenglin Road, Xuhui District, Shanghai, 200032, People's Republic of China
| | - Lu Wang
- Department of Plastic and Reconstructive Surgery, Zhongshan Hospital Fudan University, 180 Fenglin Road, Xuhui District, Shanghai, 200032, People's Republic of China
| | - Lu Lu
- Department of Plastic and Reconstructive Surgery, Zhongshan Hospital Fudan University, 180 Fenglin Road, Xuhui District, Shanghai, 200032, People's Republic of China
| | - Fa-Zhi Qi
- Department of Plastic and Reconstructive Surgery, Zhongshan Hospital Fudan University, 180 Fenglin Road, Xuhui District, Shanghai, 200032, People's Republic of China.
| |
Collapse
|
11
|
Kwaffo YA, Sarpong-Duah M, Owusu-Boateng K, Gbewonyo WS, Adjimani JP, Mosi L. Natural antioxidants attenuate mycolactone toxicity to RAW 264.7 macrophages. Exp Biol Med (Maywood) 2021; 246:1884-1894. [PMID: 34038223 DOI: 10.1177/15353702211015628] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022] Open
Abstract
Mycobacterium ulcerans produces a macrolide exotoxin, mycolactone which suppresses immune cells activity, is toxic to most cells and the key virulence factor in the pathogenesis of Buruli ulcer disease. Mycolactone is reported to mediate the production of reactive oxygen species in keratinocytes; cells that play critical role in wound healing. Increased levels of reactive oxygen species have been shown to disrupt the well-ordered process of wound repair; hence, the function of wound-healing cells such as macrophages, keratinocytes, and fibroblast could be impaired in the presence of the reactive oxygen species mediator, mycolactone. To ensure regeneration of tissues in chronic ulcers, with proper and timely healing of the wounds, natural antioxidants that can combat the effects of induced reactive oxygen species in wound-healing cells ought to be investigated. Reactive oxygen species activity was determined in mycolactone-treated RAW 264.7 macrophages and the scavenging ability of the antioxidants (ascorbic acid, gallic acid, and green tea kombucha) against mycolactone-induced reactive oxygen species (superoxide anions) was assessed using fluorescein probe (DCF-DA) and nitroblue tetrazolium dye. Cytotoxicity of the antioxidants, mycolactone, and the protective effect of the antioxidants on the cells upon treatment with mycolactone were determined using the Alamar blue assay. The expression levels of endogenous antioxidant enzyme genes (superoxide dismutase, catalase, and glutathione peroxidase) in response to mycolactone-mediated reactive oxygen species were determined using RT-qPCR. Mycolactone induced the production of reactive oxygen species in RAW 264.7 macrophages, and the resulting superoxide anions were scavenged by some of the antioxidants. The selected endogenous antioxidant enzyme genes in the macrophages were upregulated in the presence of the antioxidants and mycolactone. The exogenously supplied ascorbic acid and green tea kombucha offered moderate protection to the macrophages against the toxicity of mycolactone. We conclude that the results provide insights into alternate and adjunct therapeutic approaches in Buruli ulcer treatment, which could significantly attenuate the toxicity of the pathogenic factor; mycolactone.
Collapse
Affiliation(s)
- Yvonne A Kwaffo
- West African Centre for Cell Biology of Infectious Disease (WACCBIP), Department of Biochemistry, Cell and Molecular Biology, University of Ghana, Accra 0000, Ghana.,Department of Biochemistry, Cell and Molecular Biology, University of Ghana, Accra 0000, Ghana
| | - Mabel Sarpong-Duah
- Department of Biochemistry, Cell and Molecular Biology, University of Ghana, Accra 0000, Ghana
| | - Kwabena Owusu-Boateng
- West African Centre for Cell Biology of Infectious Disease (WACCBIP), Department of Biochemistry, Cell and Molecular Biology, University of Ghana, Accra 0000, Ghana.,Department of Biochemistry, Cell and Molecular Biology, University of Ghana, Accra 0000, Ghana
| | - Winfred Sk Gbewonyo
- Department of Biochemistry, Cell and Molecular Biology, University of Ghana, Accra 0000, Ghana
| | - Jonathan P Adjimani
- Department of Biochemistry, Cell and Molecular Biology, University of Ghana, Accra 0000, Ghana
| | - Lydia Mosi
- West African Centre for Cell Biology of Infectious Disease (WACCBIP), Department of Biochemistry, Cell and Molecular Biology, University of Ghana, Accra 0000, Ghana.,Department of Biochemistry, Cell and Molecular Biology, University of Ghana, Accra 0000, Ghana
| |
Collapse
|
12
|
Effect of Pig-Adipose-Derived Stem Cells' Conditioned Media on Skin Wound-Healing Characteristics In Vitro. Int J Mol Sci 2021; 22:ijms22115469. [PMID: 34067360 PMCID: PMC8196863 DOI: 10.3390/ijms22115469] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2021] [Revised: 05/17/2021] [Accepted: 05/19/2021] [Indexed: 01/05/2023] Open
Abstract
The primary mechanism by which adipose-derived stem cells (ASCs) exert their reparative or regenerative potential relies predominantly on paracrine action. Secretory abilities of ASCs have been found to be amplified by hypoxia pre-conditioning. This study investigates the impact of hypoxia (1% O2) on the secretome composition of pig ASCs (pASCs) and explores the effect of pASCs’ conditioned media (CM) on skin cell functions in vitro and the expression of markers attributed to wound healing. Exposure of pASCs to hypoxia increased levels of vascular endothelial growth factor (VEGF) in CM-Hyp compared to CM collected from the cells cultured in normoxia (CM-Nor). CM-Hyp promoted the migratory ability of pig keratinocytes (pKERs) and delayed migration of pig dermal fibroblasts (pDFs). Exposure of pKERs to either CM-Nor or CM-Hyp decreased the levels of pro-fibrotic indicators WNT10A and WNT11. Furthermore, CM-Hyp enhanced the expression of KRT14, the marker of the basal epidermis layer. In contrast, CM-Nor showed a stronger effect on pDFs manifested by increases in TGFB1, COL1A1, COL3A1, and FN1 mRNA expression. The formation of three-dimensional endothelial cell networks was improved in the presence of CM-Hyp. Overall, our results demonstrate that the paracrine activity of pASCs affects skin cells, and this property might be used to modulate wound healing.
Collapse
|
13
|
Macrophage recruitment in immune-privileged lens during capsule repair, necrotic fiber removal, and fibrosis. iScience 2021; 24:102533. [PMID: 34142044 PMCID: PMC8188486 DOI: 10.1016/j.isci.2021.102533] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2021] [Revised: 04/01/2021] [Accepted: 05/10/2021] [Indexed: 12/26/2022] Open
Abstract
Emerging evidence challenges the lens as an immune-privileged organ. Here, we provide a direct mechanism supporting a role of macrophages in lens capsule rupture repair. Posterior lens capsule rupture in a connexin 50 and aquaporin 0 double-knockout mouse model resulted in lens tissue extrusion into the vitreous cavity with formation of a “tail-like” tissue containing delayed regressed hyaloid vessels, fibrotic tissue and macrophages at postnatal (P) 15 days. The macrophages declined after P 30 days with M2 macrophages detected inside the lens. By P 90 days, the “tail-like” tissue completely disappeared and the posterior capsule rupture was sealed with thick fibrotic tissue. Colony-stimulating factor 1 (CSF-1) accelerated capsule repair, whereas inhibition of the CSF-1 receptor delayed the repair. Together, these results suggest that lens posterior rupture leads to the recruitment of macrophages delivered by the regression delayed hyaloid vessels. CSF-1-activated M2 macrophages mediate capsule rupture repair and development of fibrosis. Lens posterior rupture delays regression of the hyaloid vessels. Lens posterior rupture recruits macrophages delivered by the hyaloid vessels. Macrophages mediate necrotic fiber cell removal and capsule rupture sealing. CSF-1 activated M2 macrophages facilitate capsular rupture sealing by fibrosis.
Collapse
|
14
|
Zannotti A, Greco S, Pellegrino P, Giantomassi F, Delli Carpini G, Goteri G, Ciavattini A, Ciarmela P. Macrophages and Immune Responses in Uterine Fibroids. Cells 2021; 10:cells10050982. [PMID: 33922329 PMCID: PMC8146588 DOI: 10.3390/cells10050982] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2021] [Revised: 04/19/2021] [Accepted: 04/20/2021] [Indexed: 12/12/2022] Open
Abstract
Uterine fibroids represent the most common benign tumors of the uterus. They are considered a typical fibrotic disorder. In fact, the extracellular matrix (ECM) proteins—above all, collagen 1A1, fibronectin and versican—are upregulated in this pathology. The uterine fibroids etiology has not yet been clarified, and this represents an important matter about their resolution. A model has been proposed according to which the formation of an altered ECM could be the result of an excessive wound healing, in turn driven by a dysregulated inflammation process. A lot of molecules act in the complex inflammatory response. Macrophages have a great flexibility since they can assume different phenotypes leading to the tissue repair process. The dysregulation of macrophage proliferation, accumulation and infiltration could lead to an uncontrolled tissue repair and to the consequent pathological fibrosis. In addition, molecules such as monocyte chemoattractant protein-1 (MCP-1), granulocyte macrophage-colony-stimulating factor (GM-CSF), transforming growth factor-beta (TGF-β), activin A and tumor necrosis factor-alfa (TNF-α) were demonstrated to play an important role in the macrophage action within the uncontrolled tissue repair that contributes to the pathological fibrosis that represents a typical feature of the uterine fibroids.
Collapse
Affiliation(s)
- Alessandro Zannotti
- Department of Specialist and Odontostomatological Clinical Sciences, Università Politecnica delle Marche, 60126 Ancona, Italy; (A.Z.); (G.D.C.); (A.C.)
- Department of Experimental and Clinical Medicine, Università Politecnica delle Marche, 60126 Ancona, Italy; (S.G.); (P.P.)
| | - Stefania Greco
- Department of Experimental and Clinical Medicine, Università Politecnica delle Marche, 60126 Ancona, Italy; (S.G.); (P.P.)
| | - Pamela Pellegrino
- Department of Experimental and Clinical Medicine, Università Politecnica delle Marche, 60126 Ancona, Italy; (S.G.); (P.P.)
| | - Federica Giantomassi
- Department of Biomedical Sciences and Public Health, Università Politecnica delle Marche, 60126 Ancona, Italy; (F.G.); (G.G.)
| | - Giovanni Delli Carpini
- Department of Specialist and Odontostomatological Clinical Sciences, Università Politecnica delle Marche, 60126 Ancona, Italy; (A.Z.); (G.D.C.); (A.C.)
| | - Gaia Goteri
- Department of Biomedical Sciences and Public Health, Università Politecnica delle Marche, 60126 Ancona, Italy; (F.G.); (G.G.)
| | - Andrea Ciavattini
- Department of Specialist and Odontostomatological Clinical Sciences, Università Politecnica delle Marche, 60126 Ancona, Italy; (A.Z.); (G.D.C.); (A.C.)
| | - Pasquapina Ciarmela
- Department of Experimental and Clinical Medicine, Università Politecnica delle Marche, 60126 Ancona, Italy; (S.G.); (P.P.)
- Correspondence: ; Tel.:+39-071-220-6270
| |
Collapse
|
15
|
Immunomodulatory biomaterials and their application in therapies for chronic inflammation-related diseases. Acta Biomater 2021; 123:1-30. [PMID: 33484912 DOI: 10.1016/j.actbio.2021.01.025] [Citation(s) in RCA: 43] [Impact Index Per Article: 14.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2020] [Revised: 12/05/2020] [Accepted: 01/15/2021] [Indexed: 02/06/2023]
Abstract
The degree of tissue injuries such as the level of scarring or organ dysfunction, and the immune response against them primarily determine the outcome and speed of healing process. The successful regeneration of functional tissues requires proper modulation of inflammation-producing immune cells and bioactive factors existing in the damaged microenvironment. In the tissue repair and regeneration processes, different types of biomaterials are implanted either alone or by combined with other bioactive factors, which will interact with the immune systems including immune cells, cytokines and chemokines etc. to achieve different results highly depending on this interplay. In this review article, the influences of different types of biomaterials such as nanoparticles, hydrogels and scaffolds on the immune cells and the modification of immune-responsive factors such as reactive oxygen species (ROS), cytokines, chemokines, enzymes, and metalloproteinases in tissue microenvironment are summarized. In addition, the recent advances of immune-responsive biomaterials in therapy of inflammation-associated diseases such as myocardial infarction, spinal cord injury, osteoarthritis, inflammatory bowel disease and diabetic ulcer are discussed.
Collapse
|
16
|
Hirman AR, Du L, Cheng S, Zheng H, Duo L, Zhai Q, Xu J. MiR-133a-3p inhibits scar formation in scalded mice and suppresses the proliferation and migration of scar derived-fibroblasts by targeting connective tissue growth factor. Exp Anim 2021; 70:322-332. [PMID: 33658464 PMCID: PMC8390314 DOI: 10.1538/expanim.20-0159] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/10/2023] Open
Abstract
Excessive scar formation post burn injury can cause great pain to the patients. MiR-133a-3p has been demonstrated to be anti-fibrotic in some fibrosis-related
diseases. However, its possible role in scar formation has not been elucidated yet. In present study, the effect of miR-133a-3p on scar formation was
investigated in a scalded model of mice. Moreover, the function of miR-133a-3p on proliferation and migration of scar-derived fibroblasts (SFs) was studied
in vitro. It was found that miR-133a-3p was dramatically downregulated in scar tissue of scalded mice. Upregulation of miR-133a-3p by
miR-133a-3p agomir obviously inhibited the scar formation in scalded mice. Histological staining showed that upregulation of miR-133a-3p attenuated the
excessive deposition of collagen in scar tissue of scalded mice. In vitro study showed that upregulation of miR-133a-3p effectively suppressed
the proliferation and migration of SFs. Besides, upregulation of miR-133a-3p attenuated the protein levels of α-smooth muscle actin (α-SMA) and collagen I,
indicating that miR-133a-3p could suppress the activation of SFs. The expression of connective tissue growth factor (CTGF), a critical mediator in cell
proliferation, migration and extracellular matrix (ECM) synthesis, was also downregulated by the upregulation of miR-133a-3p. Luciferase reporter assay
validated that CTGF was directly targeted by miR-133a-3p. In addition, overexpression of CTGF abolished the effect of miR-133a-3p on inhibiting the
proliferation, migration and activation of SFs, indicating that miR-133a-3p functioned by targeting CTGF. Therefore, miR-133a-3p might be a promising target for
treating pathological scars.
Collapse
Affiliation(s)
- Abdul Razaq Hirman
- Department of Dermatology, Shengjing Hospital of China Medical University, No. 36, Sanhao Street, Shenyang 110004, P.R. China
| | - Lili Du
- Department of Pathophysiology, College of Basic Medical Science, China Medical University, No. 77, Puhe Road, Shenbei New District, Shenyang 110122, P.R. China
| | - Shaohang Cheng
- Department of Dermatology, Shengjing Hospital of China Medical University, No. 36, Sanhao Street, Shenyang 110004, P.R. China
| | - Heng Zheng
- Department of Dermatology, Central Hospital Affiliated to Shenyang Medical College, No. 7, Nanqi West Road, Tiexi District, Shenyang 110024, P.R. China
| | - Linna Duo
- Department of Dermatology, Shengjing Hospital of China Medical University, No. 36, Sanhao Street, Shenyang 110004, P.R. China
| | - Qianyu Zhai
- Department of Dermatology, Shengjing Hospital of China Medical University, No. 36, Sanhao Street, Shenyang 110004, P.R. China
| | - Jing Xu
- Department of Dermatology, Shengjing Hospital of China Medical University, No. 36, Sanhao Street, Shenyang 110004, P.R. China
| |
Collapse
|
17
|
Qian S, Tang Y, Tang QQ. Adipose tissue plasticity and the pleiotropic roles of BMP signaling. J Biol Chem 2021; 296:100678. [PMID: 33872596 PMCID: PMC8131923 DOI: 10.1016/j.jbc.2021.100678] [Citation(s) in RCA: 20] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2020] [Revised: 04/11/2021] [Accepted: 04/15/2021] [Indexed: 12/15/2022] Open
Abstract
Adipose tissues, including white, beige, and brown adipose tissue, have evolved to be highly dynamic organs. Adipose tissues undergo profound changes during development and regeneration and readily undergo remodeling to meet the demands of an everchanging metabolic landscape. The dynamics are determined by the high plasticity of adipose tissues, which contain various cell types: adipocytes, immune cells, endothelial cells, nerves, and fibroblasts. There are numerous proteins that participate in regulating the plasticity of adipose tissues. Among these, bone morphogenetic proteins (BMPs) were initially found to regulate the differentiation of adipocytes, and they are being reported to have pleiotropic functions by emerging studies. Here, in the first half of the article, we summarize the plasticity of adipocytes and macrophages, which are two groups of cells targeted by BMP signaling in adipose tissues. We then review how BMPs regulate the differentiation, death, and lipid metabolism of adipocytes. In addition, the potential role of BMPs in regulating adipose tissue macrophages is considered. Finally, the expression of BMPs in adipose tissues and their metabolic relevance are discussed.
Collapse
Affiliation(s)
- Shuwen Qian
- The Key Laboratory of Metabolism and Molecular Medicine of the Ministry of Education, Department of Biochemistry and Molecular Biology of School of Basic Medical Sciences, and Department of Endocrinology and Metabolism of Zhongshan Hospital, Fudan University, Shanghai, China
| | - Yan Tang
- The Key Laboratory of Metabolism and Molecular Medicine of the Ministry of Education, Department of Biochemistry and Molecular Biology of School of Basic Medical Sciences, and Department of Endocrinology and Metabolism of Zhongshan Hospital, Fudan University, Shanghai, China
| | - Qi-Qun Tang
- The Key Laboratory of Metabolism and Molecular Medicine of the Ministry of Education, Department of Biochemistry and Molecular Biology of School of Basic Medical Sciences, and Department of Endocrinology and Metabolism of Zhongshan Hospital, Fudan University, Shanghai, China.
| |
Collapse
|
18
|
He YS, Yang XK, Hu YQ, Xiang K, Pan HF. Emerging role of Fli1 in autoimmune diseases. Int Immunopharmacol 2020; 90:107127. [PMID: 33234418 DOI: 10.1016/j.intimp.2020.107127] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2020] [Revised: 09/21/2020] [Accepted: 10/19/2020] [Indexed: 11/25/2022]
Abstract
The Ets transcription factor family exerts crucial role in cell proliferation, apoptosis, differentiation and migration. Friend leukemia integration 1 (Fli1), a member of the Ets family, is expressed in fibroblasts, endothelial cells and immune cells. Fli1 gene is participated in the development, proliferation, activation, migration and other processes of immune cells. Fli1 can also affect the function of immune cells by regulating cytokines and chemokines. Emerging evidence has shown that Fli1 is implicated in the etiology of several autoimmune diseases, including systemic sclerosis (SSc) and systemic lupus erythematosus (SLE). In this review, we mainly discuss the current evidence for the role of Fli1 in these diseases.
Collapse
Affiliation(s)
- Yi-Sheng He
- Department of Epidemiology and Biostatistics, School of Public Health, Anhui Medical University, 81 Meishan Road, Hefei, Anhui, China; Inflammation and Immune Mediated Diseases Laboratory of Anhui Province, 81 Meishan Road, Hefei, Anhui, China
| | - Xiao-Ke Yang
- Department of Rheumatology and Immunology, the First Affiliated Hospital of Anhui Medical University, 218 Jixi Road, Hefei, Anhui, China
| | - Yu-Qian Hu
- Department of Epidemiology and Biostatistics, School of Public Health, Anhui Medical University, 81 Meishan Road, Hefei, Anhui, China; Inflammation and Immune Mediated Diseases Laboratory of Anhui Province, 81 Meishan Road, Hefei, Anhui, China
| | - Kun Xiang
- Department of Epidemiology and Biostatistics, School of Public Health, Anhui Medical University, 81 Meishan Road, Hefei, Anhui, China; Inflammation and Immune Mediated Diseases Laboratory of Anhui Province, 81 Meishan Road, Hefei, Anhui, China
| | - Hai-Feng Pan
- Department of Epidemiology and Biostatistics, School of Public Health, Anhui Medical University, 81 Meishan Road, Hefei, Anhui, China; Inflammation and Immune Mediated Diseases Laboratory of Anhui Province, 81 Meishan Road, Hefei, Anhui, China.
| |
Collapse
|
19
|
Rüger BM, Buchacher T, Dauber EM, Pasztorek M, Uhrin P, Fischer MB, Breuss JM, Leitner GC. De novo Vessel Formation Through Cross-Talk of Blood-Derived Cells and Mesenchymal Stromal Cells in the Absence of Pre-existing Vascular Structures. Front Bioeng Biotechnol 2020; 8:602210. [PMID: 33330432 PMCID: PMC7718010 DOI: 10.3389/fbioe.2020.602210] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2020] [Accepted: 10/26/2020] [Indexed: 12/31/2022] Open
Abstract
BACKGROUND The generation of functional blood vessels remains a key challenge for regenerative medicine. Optimized in vitro culture set-ups mimicking the in vivo perivascular niche environment during tissue repair may provide information about the biological function and contribution of progenitor cells to postnatal vasculogenesis, thereby enhancing their therapeutic potential. AIM We established a fibrin-based xeno-free human 3D in vitro vascular niche model to study the interaction of mesenchymal stromal cells (MSC) with peripheral blood mononuclear cells (PBMC) including circulating progenitor cells in the absence of endothelial cells (EC), and to investigate the contribution of this cross-talk to neo-vessel formation. MATERIALS AND METHODS Bone marrow-derived MSC were co-cultured with whole PBMC, enriched monocytes (Mo), enriched T cells, and Mo together with T cells, respectively, obtained from leukocyte reduction chambers generated during the process of single-donor platelet apheresis. Cells were embedded in 3D fibrin matrices, using exclusively human-derived culture components without external growth factors. Cytokine secretion was analyzed in supernatants of 3D cultures by cytokine array, vascular endothelial growth factor (VEGF) secretion was quantified by ELISA. Cellular and structural re-arrangements were characterized by immunofluorescence and confocal laser-scanning microscopy of topographically intact 3D fibrin gels. RESULTS 3D co-cultures of MSC with PBMC, and enriched Mo together with enriched T cells, respectively, generated, within 2 weeks, complex CD31+/CD34+ vascular structures, surrounded by basement membrane collagen type-IV+ cells and matrix, in association with increased VEGF secretion. PBMC contained CD31+CD34+CD45dimCD14- progenitor-type cells, and EC of neo-vessels were PBMC-derived. Vascular structures showed intraluminal CD45+ cells that underwent apoptosis thereby creating a lumen. Cross-talk of MSC with enriched Mo provided a pro-angiogenic paracrine environment. MSC co-cultured with enriched T cells formed "cell-in-cell" structures generated through internalization of T cells by CD31+CD45 dim/ - cells. No vascular structures were detected in co-cultures of MSC with either Mo or T cells. CONCLUSION Our xeno-free 3D in vitro vascular niche model demonstrates that a complex synergistic network of cellular, extracellular and paracrine cross-talk can contribute to de novo vascular development through self-organization via co-operation of immune cells with blood-derived progenitor cells and MSC, and thereby may open a new perspective for advanced vascular tissue engineering in regenerative medicine.
Collapse
Affiliation(s)
- Beate M. Rüger
- Department of Blood Group Serology and Transfusion Medicine, Medical University of Vienna, Vienna, Austria
| | - Tanja Buchacher
- Turku Bioscience Centre, University of Turku and Åbo Akademi University, Turku, Finland
| | - Eva-Maria Dauber
- Department of Blood Group Serology and Transfusion Medicine, Medical University of Vienna, Vienna, Austria
| | - Markus Pasztorek
- Department of Health Sciences, Medicine and Research, Faculty of Health and Medicine, Danube University Krems, Krems an der Donau, Austria
| | - Pavel Uhrin
- Department of Vascular Biology and Thrombosis Research, Center for Physiology and Pharmacology, Medical University of Vienna, Vienna, Austria
| | - Michael B. Fischer
- Department of Blood Group Serology and Transfusion Medicine, Medical University of Vienna, Vienna, Austria
- Department of Health Sciences, Medicine and Research, Faculty of Health and Medicine, Danube University Krems, Krems an der Donau, Austria
| | - Johannes M. Breuss
- Department of Vascular Biology and Thrombosis Research, Center for Physiology and Pharmacology, Medical University of Vienna, Vienna, Austria
| | - Gerda C. Leitner
- Department of Blood Group Serology and Transfusion Medicine, Medical University of Vienna, Vienna, Austria
| |
Collapse
|
20
|
Hachim D, LoPresti ST, Rege RD, Umeda Y, Iftikhar A, Nolfi AL, Skillen CD, Brown BN. Distinct macrophage populations and phenotypes associated with IL-4 mediated immunomodulation at the host implant interface. Biomater Sci 2020; 8:5751-5762. [PMID: 32945303 PMCID: PMC7641101 DOI: 10.1039/d0bm00568a] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/28/2022]
Abstract
The host macrophage response to implants has shown to be affected by tissue location and physio-pathological conditions of the patient. Success in immunomodulatory strategies is thus predicated on the proper understanding of the macrophage populations participating on each one of these contexts. The present study uses an in vivo implantation model to analyze how immunomodulation via an IL-4 eluting implant affects distinct macrophage populations at the tissue-implant interface and how this may affect downstream regenerative processes. Populations identified as F4/80+, CD68+ and CD11b+ macrophages at the peri-implant space showed distinct susceptibility to polarize towards an M2-like phenotype under the effects of delivered IL-4. Also, the presence of the coating resulted in a significant reduction in F4/80+ macrophages, while other populations remained unchanged. These results suggests that the F4/80+ macrophage population may be predominant in the early stages of the host response at the surface of these implants, in contrast to CD11b+ macrophage populations which were either fewer in number or located more distant from the implant surface. Gene expression assays showed increased proteolytic activity and diminished matrix deposition as possible mechanisms explaining the decreased fibrotic capsule deposition and improved peri-implant tissue quality shown in previous studies using IL-4 eluting coatings. The pattern of M2-like gene expression promoted by IL-4 was correlated with glycosaminoglycan production within the site of implantation at early stages of the host response, suggesting a significant role in this response. These findings demonstrate that immunomodulatory strategies can be utilized to design and implement targeted delivery for improving biomaterial performance.
Collapse
Affiliation(s)
- Daniel Hachim
- McGowan Institute for Regenerative Medicine, University of Pittsburgh, 450 Technology Drive, Suite 300, Pittsburgh, PA 15219, USA.
| | | | | | | | | | | | | | | |
Collapse
|
21
|
Moysenovich AM, Moisenovich MM, Sudina AK, Tatarskiy VV, Khamidullina AI, Yastrebova MA, Davydova LI, Bogush VG, Debabov VG, Arkhipova AY, Shaitan KV, Shtil AA, Demina IA. Recombinant Spidroin Films Attenuate Individual Markers of Glucose Induced Aging in NIH 3T3 Fibroblasts. BIOCHEMISTRY (MOSCOW) 2020; 85:808-819. [PMID: 33040725 DOI: 10.1134/s0006297920070093] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
The effect of bioresorbable materials on aging in cultured mouse NIH 3T3 fibroblasts treated with elevated glucose concentration was investigated. The cells were grown on films produced from the silkworm fibroin and rS1/9, a recombinant analog of Nephila clavipes spidroin 1. Exposure to 50 mM glucose of the cells grown on uncoated glass support resulted in the cell growth retardation. The average areas of the cells and nuclei and the percentage of apoptotic cells increased, whereas the amount of soluble collagen decreased. In contrast, on the fibroin and spidroin films, the cell density and the percentage of 5-bromo-2'-deoxyuridine (BrdU)-positive cells were higher vs. the cells grown on the glass support. The films protected NIH 3T3 fibroblasts from the glucose-induced death. The most prominent effects on the cell density, BrdU incorporation, and apoptosis prevention were observed in the cells cultured on spidroin films. Unlike the cells grown on glass support (decrease in the soluble collagen production) or fibroin (no effect), production of soluble collagen by the cells grown on spidroin films increased after cell exposure to 50 mM glucose. Molecular analysis demonstrated that 50 mM glucose upregulated phosphorylation of the NFκB heterodimer p65 subunit in the cells grown on the glass support. The treatment of cells grown on fibroin films with 5.5 mM or 50 mM glucose had no effect on p65 phosphorylation. The same treatment decreased p65 phosphorylation in the cells on the spidroin films. These results demonstrate the anti-aging efficacy of biomaterials derived from the silk proteins and suggest that spidroin is more advantageous for tissue engineering and therapy than fibroin.
Collapse
Affiliation(s)
- A M Moysenovich
- Faculty of Biology, Lomonosov Moscow State University, Moscow, 119234, Russia
| | - M M Moisenovich
- Faculty of Biology, Lomonosov Moscow State University, Moscow, 119234, Russia.
| | - A K Sudina
- Faculty of Biology, Lomonosov Moscow State University, Moscow, 119234, Russia
| | - V V Tatarskiy
- Blokhin National Medical Research Center of Oncology, Moscow, 115478, Russia.,Institute of Gene Biology, Russian Academy of Sciences, Moscow, 119334, Russia.,National University of Science and Technology "MISiS", Moscow, 119049, Russia
| | - A I Khamidullina
- Institute of Gene Biology, Russian Academy of Sciences, Moscow, 119334, Russia
| | - M A Yastrebova
- Institute of Gene Biology, Russian Academy of Sciences, Moscow, 119334, Russia
| | - L I Davydova
- NRC "Kurchatov Institute" - GOSNIIGENETIKA, Moscow, 117519, Russia.,NRC "Kurchatov Institute", Moscow, 123182, Russia
| | - V G Bogush
- NRC "Kurchatov Institute" - GOSNIIGENETIKA, Moscow, 117519, Russia.,NRC "Kurchatov Institute", Moscow, 123182, Russia
| | - V G Debabov
- NRC "Kurchatov Institute" - GOSNIIGENETIKA, Moscow, 117519, Russia.,NRC "Kurchatov Institute", Moscow, 123182, Russia
| | - A Yu Arkhipova
- Faculty of Biology, Lomonosov Moscow State University, Moscow, 119234, Russia.,Moscow Regional Research and Clinical Institute (MONIKI), Moscow, 129110, Russia
| | - K V Shaitan
- Faculty of Biology, Lomonosov Moscow State University, Moscow, 119234, Russia.,Semenov Institute of Chemical Physics, Russian Academy of Sciences, Moscow, 119991, Russia
| | - A A Shtil
- Blokhin National Medical Research Center of Oncology, Moscow, 115478, Russia.,Institute of Gene Biology, Russian Academy of Sciences, Moscow, 119334, Russia
| | - I A Demina
- Faculty of Biology, Lomonosov Moscow State University, Moscow, 119234, Russia.,Dmitry Rogachev National Medical Research Center of Pediatric Hematology, Oncology and Immunology, Moscow, 117198, Russia
| |
Collapse
|
22
|
Bujor AM, El Adili F, Parvez A, Marden G, Trojanowska M. Fli1 Downregulation in Scleroderma Myeloid Cells Has Profibrotic and Proinflammatory Effects. Front Immunol 2020; 11:800. [PMID: 32508810 PMCID: PMC7248379 DOI: 10.3389/fimmu.2020.00800] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2020] [Accepted: 04/07/2020] [Indexed: 12/17/2022] Open
Abstract
Scleroderma (SSc) is an autoimmune connective tissue disease characterized by immune dysregulation, vasculopathy, and fibrosis. We have previously demonstrated that low Fli1 expression in SSc fibroblasts and endothelial cells plays an important role in SSc pathogenesis. Cells of myeloid and lymphoid origin also express Fli1 and are dysregulated in patients with SSc, playing key roles in disease pathogenesis. However, the role for immune Fli1 in SSc is not yet clear. Our aim was to elucidate whether Fli1 contributes to the immune dysregulation seen in SSc. Comparison of the expression of Fli1 in monocytes, B- and T-cell fractions of PBMCs isolated from SSc patients and healthy controls (HC), showed an increase in Fli1 levels in monocytes. We used siRNA transfected human myeloid cells and mouse peritoneal macrophages obtained from Fli1 flox/flox LysMCre+/+ mice, and found that markers of alternative macrophage activation were increased with Fli1 deletion. Coculture of Fli1-deficient myeloid cells and primary human or mouse fibroblasts resulted in a potent induction of collagen type I, independent of TGFβ upregulation. We next analyzed global gene expression profile in response to Fli1 downregulation, to gain further insight into the molecular mechanisms of this process and to identify differentially expressed genes in myeloid cells. Of relevance to SSc, the top most upregulated pathways were hallmark IFN-γ and IFN-α response. Additionally, several genes previously linked to SSc pathogenesis and fibrosis in general were also induced, including CCL2, CCL7, MMP12, and CXCL10. ANKRD1, a profibrotic transcription co-regulator was the top upregulated gene in our array. Our results show that Fli1-deficient myeloid cells share key features with cells from SSc patients, with higher expression of profibrotic markers and activation of interferon responsive genes, thus suggesting that dysregulation of Fli1 in myeloid cells may contribute to SSc pathogenesis.
Collapse
MESH Headings
- Animals
- Autoimmune Diseases
- Cells, Cultured
- Coculture Techniques
- Disease Models, Animal
- Down-Regulation
- Fibroblasts/metabolism
- Fibrosis/metabolism
- Fibrosis/pathology
- Gene Expression
- Healthy Volunteers
- Humans
- Macrophages, Peritoneal/metabolism
- Mice
- Mice, Inbred C57BL
- Mice, Knockout
- Mice, Transgenic
- Monocytes/metabolism
- Myeloid Cells/metabolism
- Proto-Oncogene Protein c-fli-1/metabolism
- RNA, Small Interfering
- Scleroderma, Systemic/genetics
- Scleroderma, Systemic/immunology
- Scleroderma, Systemic/metabolism
- Skin/metabolism
- Transforming Growth Factor beta/metabolism
Collapse
Affiliation(s)
- Andreea M. Bujor
- Division of Rheumatology, Department of Medicine, Arthritis and Autoimmune Diseases Research Center, Boston University School of Medicine, Boston, MA, United States
| | - Fatima El Adili
- Division of Rheumatology, Department of Medicine, Arthritis and Autoimmune Diseases Research Center, Boston University School of Medicine, Boston, MA, United States
- Division of Rheumatology, Department of Medicine, Whitaker Cardiovascular Institute, Boston University School of Medicine, Boston, MA, United States
| | - Arshi Parvez
- Division of Rheumatology, Department of Medicine, Arthritis and Autoimmune Diseases Research Center, Boston University School of Medicine, Boston, MA, United States
| | - Grace Marden
- Division of Rheumatology, Department of Medicine, Arthritis and Autoimmune Diseases Research Center, Boston University School of Medicine, Boston, MA, United States
| | - Maria Trojanowska
- Division of Rheumatology, Department of Medicine, Arthritis and Autoimmune Diseases Research Center, Boston University School of Medicine, Boston, MA, United States
| |
Collapse
|
23
|
Ishikawa M, Yamamoto T. Antifibrogenic effects of C-C chemokine receptor type 2 antagonist in a bleomycin-induced scleroderma model. Exp Dermatol 2020; 30:179-184. [PMID: 32096250 DOI: 10.1111/exd.14088] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2019] [Revised: 02/06/2020] [Accepted: 02/20/2020] [Indexed: 12/29/2022]
Abstract
There have been several studies on the role of the monocyte chemotactic protein-1/C-C chemokine receptor type 2 (CCR2) signalling pathway in fibrotic diseases, which identified the blockade of this pathway as a potential therapeutic target for treating fibrosis. We examined the efficacy of CCR2 antagonist (RS-504393) in a mouse model of scleroderma induced by bleomycin. RS-504393 was administered via intradermal injection 6 hours prior to bleomycin injection, in the same sites. Histopathological examination showed that RS-504393 treatment suppressed dermal fibrosis and decreased dermal thickness. The numbers of mast cells and myofibroblasts in the skin of RS-504393-treated mice were significantly lower compared with those in PBS-treated mice. Moreover, the amount of collagen in the skin of RS-504393-treated mice was significantly lower compared with that in the PBS-treated mice. Additionally, mRNA levels of TGF-β1 and collagen I alpha 1 in sclerotic skin were significantly decreased by RS-504393, and semiquantitative histopathological scoring of the lungs showed inhibition of fibrosis in RS-504393-treated mice. The amount of collagen in the lung of the RS-504393-treated mice was lower compared with that in the PBS-treated mice. These data suggest that CCR2 antagonist RS-504393 may be a therapeutic agent for human scleroderma.
Collapse
Affiliation(s)
- Masato Ishikawa
- Department of Dermatology, Fukushima Medical University, Fukushima, Japan
| | - Toshiyuki Yamamoto
- Department of Dermatology, Fukushima Medical University, Fukushima, Japan
| |
Collapse
|
24
|
Recombinant HvRNASET2 protein induces marked connective tissue remodelling in the invertebrate model Hirudo verbana. Cell Tissue Res 2020; 380:565-579. [PMID: 32043208 DOI: 10.1007/s00441-020-03174-0] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2019] [Accepted: 01/20/2020] [Indexed: 01/03/2023]
Abstract
The RNASET2 ribonuclease, belonging to the highly conserved RH/T2/s RNase gene family, has been recently shown to modulate inflammatory processes in both vertebrates and invertebrates. Indeed, the RNASET2 protein acts as a chemoattractor for macrophages in both in vitro and in vivo experimental settings and its expression significantly increases following bacterial infections. Moreover, we recently observed that injection of human recombinant RNASET2 protein in the body wall of the medicinal leech (a consolidated invertebrate model for both immune response and tissue regeneration) not only induced immune cell recruitment but also apparently triggered massive connective tissue remodelling as well. Based on these data, we evaluate here a possible role of leech recombinant RNASET2 protein (rHvRNASET2) in connective tissue remodelling by characterizing the cell types involved in this process through histochemical, morphological and immunofluorescent assays. Moreover, a time-course expression analysis of newly synthesized pro-collagen1α1 (COL1α1) and basic FGF receptor (bFGFR, a known fibroblast marker) following rHvRNASET2 injection in the leech body wall further supported the occurrence of rHvRNASET2-mediated matrix remodelling. Human MRC-5 fibroblast cells were also investigated in order to evaluate their pattern of collagen neosynthesis driven by rHvRNASET2 injection.Taken together, the data reported in this work provide compelling evidence in support of a pleiotropic role for RNASET2 in orchestrating an evolutionarily conserved crosstalk between inflammatory response and regenerative process, based on macrophage recruitment and fibroblast activation, coupled to a massive extracellular reorganization.
Collapse
|
25
|
Sharma JR, Lebeko M, Kidzeru EB, Khumalo NP, Bayat A. In Vitro and Ex Vivo Models for Functional Testing of Therapeutic Anti-scarring Drug Targets in Keloids. Adv Wound Care (New Rochelle) 2019; 8:655-670. [PMID: 31827980 PMCID: PMC6904937 DOI: 10.1089/wound.2019.1040] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2019] [Accepted: 09/09/2019] [Indexed: 01/08/2023] Open
Abstract
Significance: Keloids are benign fibro-proliferative raised dermal lesions that spread beyond the original borders of the wound, continue to grow, rarely regress, and are the most common in pigmented individuals after an abnormal wound healing response. The current treatment failure and respective challenges involved highlighting the underlying issue that the etiopathogenesis of keloids is still not well understood. Disease models are required to better understand the disease pathogenesis. It is not possible to establish keloids in animals because of the uniqueness of this disease to human skin. To address this challenge, along these lines, non-animal reproducible models are vital in investigating molecular mechanisms of keloid pathogenesis and therapeutics development. Recent Advances: Various non-animal models have been developed to better understand the molecular mechanisms involved in keloid scarring and aid in identifying and evaluating the therapeutic potential of novel drug candidates. In this scenario, the current review aims at describing in vitro monocultures, co-cultures, organotypic cultures, and ex vivo whole skin keloid tissue organ culture models. Critical Issues and Future Directions: Current treatment options for keloids are far from securing a cure or preventing disease recurrence. Identifying universally accepted effective therapy for keloids has been hampered by the absence of appropriate disease model systems. Animal models do not accurately mimic the disease, thus non-animal model systems are pivotal in keloid research. The use of these models is essential not only for a better understanding of disease biology but also for identifying and evaluating novel drug targets.
Collapse
Affiliation(s)
- Jyoti R. Sharma
- Hair and Skin Research Laboratory, Division of Dermatology, Department of Medicine, Faculty of Health Sciences and Groote Schuur Hospital, University of Cape Town, Cape Town, South Africa
| | - Maribanyana Lebeko
- Hair and Skin Research Laboratory, Division of Dermatology, Department of Medicine, Faculty of Health Sciences and Groote Schuur Hospital, University of Cape Town, Cape Town, South Africa
| | - Elvis B. Kidzeru
- Hair and Skin Research Laboratory, Division of Dermatology, Department of Medicine, Faculty of Health Sciences and Groote Schuur Hospital, University of Cape Town, Cape Town, South Africa
- Centre for Medical Research, Institute of Medical Research and Medical Plant Studies, Ministry of Scientific Research and Innovation, Yaoundé, Cameroon
| | - Nonhlanhla P. Khumalo
- Hair and Skin Research Laboratory, Division of Dermatology, Department of Medicine, Faculty of Health Sciences and Groote Schuur Hospital, University of Cape Town, Cape Town, South Africa
| | - Ardeshir Bayat
- Hair and Skin Research Laboratory, Division of Dermatology, Department of Medicine, Faculty of Health Sciences and Groote Schuur Hospital, University of Cape Town, Cape Town, South Africa
- Plastic and Reconstructive Surgery Research, Division of Musculoskeletal & Dermatological Sciences, NIHR Manchester Biomedical Research Centre, Manchester Academic Health Science Centre, The University of Manchester, Manchester, United Kingdom
| |
Collapse
|
26
|
Liu D, Wang J, Zhao G, Jiang P, Song M, Ding H, Wang Z, Lv H, Hu Y. CSF1-associated decrease in endometrial macrophages may contribute to Asherman's syndrome. Am J Reprod Immunol 2019; 83:e13191. [PMID: 31536655 DOI: 10.1111/aji.13191] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2019] [Revised: 08/17/2019] [Accepted: 09/06/2019] [Indexed: 12/29/2022] Open
Abstract
PROBLEM Asherman's syndrome (AS) is characterized by endometrial fibrosis leading to intrauterine adhesions and symptoms like hypomenorrhea, infertility, and recurrent pregnancy loss. Macrophages are key regulators of inflammation, tissue repair, regeneration, and fibrosis. However, the role of macrophages in AS remains unclear. METHOD OF STUDY Endometrial biopsies of AS patients and controls were collected during the late proliferating phase of menstrual cycle. Fibrosis and proliferation markers were detected by Masson's trichrome staining and immunohistochemistry. Macrophages were examined by immunostaining and flow cytometry. The expression levels of CCL2, CSF1, CSF1R, and GM-CSF were detected by quantitative real-time polymerase chain reaction (q-PCR) and immunohistochemistry. A well-differentiated endometrial cell line Ishikawa (IK) was used for in vitro studies. Macrophages differentiating from THP-1 monocytic cells were polarized by IL-4/IL-13. Their culture supernatants (M(IL-4/13)-S) were applied to H2 O2 or bleomycin-damaged IK cells. RESULTS In AS patients, endometrial stroma was replaced by fibrous tissue and cell proliferation was reduced. Macrophages in endometrial tissue were mainly alternative activated macrophages and their number was significantly decreased in AS patients. The CSF1 expression level was reduced in AS patients. M(IL-4/13)-S promoted the growth and migration of IK cells and inhibited H2 O2 -induced apoptosis. M(IL-4/13)-S protected IK cells from bleomycin-induced fibrosis. CONCLUSION Macrophages are critical cells involved in the process of endometrial repair and fibrosis. The decreased amount of endometrial macrophages may be attributed to the reduced expression level of CSF1. Manipulation of macrophage activation/function may provide a novel therapeutic target for AS.
Collapse
Affiliation(s)
- Dan Liu
- Department of Obstetrics and Gynecology, The Affiliated Drum Tower Hospital of Nanjing University Medical School, Nanjing, China
| | - Jiali Wang
- Division of Immunology, The State Key Laboratory of Pharmaceutical Biotechnology, Medical School, Nanjing University, Nanjing, China
| | - Guangfeng Zhao
- Department of Obstetrics and Gynecology, The Affiliated Drum Tower Hospital of Nanjing University Medical School, Nanjing, China
| | - Peipei Jiang
- Department of Obstetrics and Gynecology, The Affiliated Drum Tower Hospital of Nanjing University Medical School, Nanjing, China
| | - Minmin Song
- Department of Obstetrics and Gynecology, The Affiliated Drum Tower Hospital of Nanjing University Medical School, Nanjing, China
| | - Hailin Ding
- Department of Obstetrics and Gynecology, The Affiliated Drum Tower Hospital of Nanjing University Medical School, Nanjing, China
| | - Zhiyin Wang
- Department of Obstetrics and Gynecology, The Affiliated Drum Tower Hospital of Nanjing University Medical School, Nanjing, China
| | - Haining Lv
- Department of Obstetrics and Gynecology, The Affiliated Drum Tower Hospital of Nanjing University Medical School, Nanjing, China
| | - Yali Hu
- Department of Obstetrics and Gynecology, The Affiliated Drum Tower Hospital of Nanjing University Medical School, Nanjing, China
| |
Collapse
|
27
|
Santolla MF, Vivacqua A, Lappano R, Rigiracciolo DC, Cirillo F, Galli GR, Talia M, Brunetti G, Miglietta AM, Belfiore A, Maggiolini M. GPER Mediates a Feedforward FGF2/FGFR1 Paracrine Activation Coupling CAFs to Cancer Cells toward Breast Tumor Progression. Cells 2019; 8:cells8030223. [PMID: 30866584 PMCID: PMC6468560 DOI: 10.3390/cells8030223] [Citation(s) in RCA: 44] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2019] [Revised: 03/01/2019] [Accepted: 03/04/2019] [Indexed: 12/12/2022] Open
Abstract
The FGF2/FGFR1 paracrine loop is involved in the cross-talk between breast cancer cells and components of the tumor stroma as cancer-associated fibroblasts (CAFs). By quantitative PCR (qPCR), western blot, immunofluorescence analysis, ELISA and ChIP assays, we demonstrated that 17β-estradiol (E2) and the G protein estrogen receptor (GPER) agonist G-1 induce the up-regulation and secretion of FGF2 via GPER together with the EGFR/ERK/c-fos/AP-1 signaling cascade in (ER)-negative primary CAFs. Evaluating the genetic alterations from METABRIC and TCGA datasets, we then assessed that FGFR1 is the most frequently amplified FGFRs family member and its amplification/expression associates with shorter survival rates in breast cancer patients. Therefore, in order to assess the functional FGF2/FGFR1 interplay between CAFs and breast cancer cells, we generated the FGFR1-knockout MDA-MB-231 cells using CRISPR/Cas9 genome editing strategy. Using conditioned medium from estrogen-stimulated CAFs, we established that the activation of FGF2/FGFR1 paracrine signaling triggers the expression of the connective tissue growth factor (CTGF), leading to the migration and invasion of MDA-MB-231 cells. Our findings shed new light on the role elicited by estrogens through GPER in the activation of the FGF2/FGFR1 signaling. Moreover, our findings may identify further biological targets that could be considered in innovative combination strategies halting breast cancer progression.
Collapse
Affiliation(s)
- Maria Francesca Santolla
- Department of Pharmacy, Health and Nutritional Sciences, University of Calabria, 87036 Rende, Italy.
| | - Adele Vivacqua
- Department of Pharmacy, Health and Nutritional Sciences, University of Calabria, 87036 Rende, Italy.
| | - Rosamaria Lappano
- Department of Pharmacy, Health and Nutritional Sciences, University of Calabria, 87036 Rende, Italy.
| | | | - Francesca Cirillo
- Department of Pharmacy, Health and Nutritional Sciences, University of Calabria, 87036 Rende, Italy.
| | - Giulia Raffaella Galli
- Department of Pharmacy, Health and Nutritional Sciences, University of Calabria, 87036 Rende, Italy.
| | - Marianna Talia
- Department of Pharmacy, Health and Nutritional Sciences, University of Calabria, 87036 Rende, Italy.
| | - Giuseppe Brunetti
- University of Natural Resources and Life Sciences, 1180 Vienna, Austria.
| | | | - Antonino Belfiore
- Endocrinology, Department of Clinical and Experimental Medicine, University of Catania, Garibaldi-Nesima Hospital, 95122 Catania, Italy.
| | - Marcello Maggiolini
- Department of Pharmacy, Health and Nutritional Sciences, University of Calabria, 87036 Rende, Italy.
| |
Collapse
|
28
|
Ramani K, Biswas PS. Interleukin-17: Friend or foe in organ fibrosis. Cytokine 2019; 120:282-288. [PMID: 30772195 DOI: 10.1016/j.cyto.2018.11.003] [Citation(s) in RCA: 33] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2018] [Revised: 11/02/2018] [Accepted: 11/05/2018] [Indexed: 02/07/2023]
Abstract
Fibrosis affects all vital organs accounting for a staggering 45% of deaths worldwide and no effective therapies are currently available. Unresolved inflammation triggers downstream signaling events that lead to organ fibrosis. In recent years, proinflammatory cytokine Interleukin-17 (IL-17) has been implicated in several chronic inflammatory diseases that often culminate in organ damage followed by impaired wound healing and fibrosis. In this review, we outline the contribution of the IL-17 in mediating fibrotic diseases in various organs. A comprehensive understanding of the inflammatory events, and particularly the details of IL-17 signaling in vivo, could be beneficial in designing new therapeutic or preventive approaches to treat fibrosis. Additionally, understanding organ-specific differences in IL-17 activity could lead to targeted therapies and help spare other organs from unwanted side effects.
Collapse
Affiliation(s)
- Kritika Ramani
- Division of Rheumatology and Clinical Immunology, Department of Medicine, University of Pittsburgh, Pittsburgh, PA 15261, USA
| | - Partha S Biswas
- Division of Rheumatology and Clinical Immunology, Department of Medicine, University of Pittsburgh, Pittsburgh, PA 15261, USA.
| |
Collapse
|
29
|
Henrot P, Truchetet ME, Fisher G, Taïeb A, Cario M. CCN proteins as potential actionable targets in scleroderma. Exp Dermatol 2018; 28:11-18. [PMID: 30329180 DOI: 10.1111/exd.13806] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2018] [Revised: 10/01/2018] [Accepted: 10/10/2018] [Indexed: 12/26/2022]
Abstract
Systemic sclerosis (SSc) is a complex autoimmune connective tissue disease combining inflammatory, vasculopathic and fibrotic manifestations. Skin features, which give their name to the disease and are considered as diagnostic as well as prognostic markers, have not been thoroughly investigated in terms of therapeutic targets. CCN proteins (CYR61/CCN1, CTGF/CCN2, NOV/CCN3 and WISP1-2-3 as CCN4-5-6) are a family of secreted matricellular proteins implicated in major cellular processes such as cell growth, migration, differentiation. They have already been implicated in key pathophysiological processes of SSc, namely fibrosis, vasculopathy and inflammation. In this review, we discuss the possible implication of CCN proteins in SSc pathogenesis, with a special focus on skin features, and identify the potential actionable CCN targets.
Collapse
Affiliation(s)
- Pauline Henrot
- University of Bordeaux, Inserm, BMGIC, UMR1035, Bordeaux, France.,Department of Rheumatology, National Reference Center for Rare Diseases, Bordeaux University Hospital, Bordeaux, France
| | - Marie-Elise Truchetet
- Department of Rheumatology, National Reference Center for Rare Diseases, Bordeaux University Hospital, Bordeaux, France.,University of Bordeaux, CNRS, Immunoconcept, UMR 5164, Bordeaux, France
| | - Gary Fisher
- Department of Dermatology, University of Michigan Medical School, Ann Arbor, Michigan
| | - Alain Taïeb
- University of Bordeaux, Inserm, BMGIC, UMR1035, Bordeaux, France.,Department of Dermatology, University of Michigan Medical School, Ann Arbor, Michigan
| | - Muriel Cario
- University of Bordeaux, Inserm, BMGIC, UMR1035, Bordeaux, France.,Department of Dermatology and Pediatric Dermatology, National Center for Rare Skin Disorders, Hôpital Saint André, Bordeaux, France
| |
Collapse
|
30
|
Nosenko MA, Moysenovich AM, Zvartsev RV, Arkhipova AY, Zhdanova AS, Agapov II, Vasilieva TV, Bogush VG, Debabov VG, Nedospasov SA, Moisenovich MM, Drutskaya MS. Novel Biodegradable Polymeric Microparticles Facilitate Scarless Wound Healing by Promoting Re-epithelialization and Inhibiting Fibrosis. Front Immunol 2018; 9:2851. [PMID: 30564244 PMCID: PMC6288351 DOI: 10.3389/fimmu.2018.02851] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2018] [Accepted: 11/19/2018] [Indexed: 01/20/2023] Open
Abstract
Despite decades of research, the goal of achieving scarless wound healing remains elusive. One of the approaches, treatment with polymeric microcarriers, was shown to promote tissue regeneration in various in vitro models of wound healing. The in vivo effects of such an approach are attributed to transferred cells with polymeric microparticles functioning merely as inert scaffolds. We aimed to establish a bioactive biopolymer carrier that would promote would healing and inhibit scar formation in the murine model of deep skin wounds. Here we characterize two candidate types of microparticles based on fibroin/gelatin or spidroin and show that both types increase re-epithelialization rate and inhibit scar formation during skin wound healing. Interestingly, the effects of these microparticles on inflammatory gene expression and cytokine production by macrophages, fibroblasts, and keratinocytes are distinct. Both types of microparticles, as well as their soluble derivatives, fibroin and spidroin, significantly reduced the expression of profibrotic factors Fgf2 and Ctgf in mouse embryonic fibroblasts. However, only fibroin/gelatin microparticles induced transient inflammatory gene expression and cytokine production leading to an influx of inflammatory Ly6C+ myeloid cells to the injection site. The ability of microparticle carriers of equal proregenerative potential to induce inflammatory response may allow their subsequent adaptation to treatment of wounds with different bioburden and fibrotic content.
Collapse
Affiliation(s)
- Maxim A Nosenko
- Engelhardt Institute of Molecular Biology, Russian Academy of Sciences, Moscow, Russia.,Department of Biology, Lomonosov Moscow State University, Moscow, Russia
| | | | - Ruslan V Zvartsev
- Engelhardt Institute of Molecular Biology, Russian Academy of Sciences, Moscow, Russia
| | - Anastasia Y Arkhipova
- Department of Biology, Lomonosov Moscow State University, Moscow, Russia.,Moscow Regional Research and Clinical Institute ("MONIKI"), Moscow, Russia
| | - Anastasia S Zhdanova
- Engelhardt Institute of Molecular Biology, Russian Academy of Sciences, Moscow, Russia.,Department of Biology, Lomonosov Moscow State University, Moscow, Russia
| | - Igor I Agapov
- V. I. Shumakov National Medical Research Center of Transplantology and Artificial Organs, Moscow, Russia
| | - Tamara V Vasilieva
- Department of Biology, Lomonosov Moscow State University, Moscow, Russia
| | - Vladimir G Bogush
- State Research Institute for Genetics and Selection of Industrial Microorganisms of National Research Center "Kurchatov Institute", Moscow, Russia
| | - Vladimir G Debabov
- State Research Institute for Genetics and Selection of Industrial Microorganisms of National Research Center "Kurchatov Institute", Moscow, Russia
| | - Sergei A Nedospasov
- Engelhardt Institute of Molecular Biology, Russian Academy of Sciences, Moscow, Russia.,Department of Biology, Lomonosov Moscow State University, Moscow, Russia
| | | | - Marina S Drutskaya
- Engelhardt Institute of Molecular Biology, Russian Academy of Sciences, Moscow, Russia.,Department of Biology, Lomonosov Moscow State University, Moscow, Russia
| |
Collapse
|
31
|
Ramazani Y, Knops N, Elmonem MA, Nguyen TQ, Arcolino FO, van den Heuvel L, Levtchenko E, Kuypers D, Goldschmeding R. Connective tissue growth factor (CTGF) from basics to clinics. Matrix Biol 2018; 68-69:44-66. [DOI: 10.1016/j.matbio.2018.03.007] [Citation(s) in RCA: 130] [Impact Index Per Article: 21.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2017] [Revised: 03/05/2018] [Accepted: 03/06/2018] [Indexed: 02/07/2023]
|
32
|
He C, Yang Z, Jin Y, Qi X, Chu J, Deng X. ADM Scaffolds Generate a Pro-regenerative Microenvironment During Full-Thickness Cutaneous Wound Healing Through M2 Macrophage Polarization via Lamtor1. Front Physiol 2018; 9:657. [PMID: 29915541 PMCID: PMC5994424 DOI: 10.3389/fphys.2018.00657] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2018] [Accepted: 05/14/2018] [Indexed: 12/21/2022] Open
Abstract
Adult mammalian skin has a defective regenerative capacity following full-thickness cutaneous injury; this defect overshadows the complete physiological functions of the skin. Immune-mediated skin reconstruction driven by biological scaffolds is a recently developed innovative repair strategy to support regenerative wound healing. However, to date, little is known about how biological scaffolds orchestrate the immune response to promote regeneration. Here, using acellular dermal matrix (ADM) scaffolds, we discovered that the default pro-inflammatory response was altered in response to a pro-regenerative response characterized by specific M2 polarization. M2 macrophages subsequently produced a series of wound healing factors, including matrix metalloproteinases (Mmps), and growth factors which promoted cell proliferation, stabilized angiogenesis, and remodeled the extracellular matrix. Our investigations further revealed that the M2 polarization of macrophages arose from an ADM scaffold-derived amino acid sufficiency signal by collagen degradation via macrophage phagocytosis, which activated the acid-sensing pathway (v-ATPase, Lamtor1, and mTORC1). Lamtor1, the acid-sensing pathway-associated lysosomal adaptor protein was critical for inducing M2 polarization, while with the presence of extracellular interleukin 4 (IL4). Our results suggest that ADM scaffolds generate a pro-regenerative microenvironment during full-thickness cutaneous wound healing through M2 macrophage polarization via Lamtor1.
Collapse
Affiliation(s)
- Chengmin He
- MOE Key Laboratory of Laser Life Science, College of Biophotonics, South China Normal University, Guangzhou, China
| | - Zhi Yang
- MOE Key Laboratory of Laser Life Science, College of Biophotonics, South China Normal University, Guangzhou, China
| | - Ying Jin
- MOE Key Laboratory of Laser Life Science, College of Biophotonics, South China Normal University, Guangzhou, China
| | - Xiaoyang Qi
- MOE Key Laboratory of Laser Life Science, College of Biophotonics, South China Normal University, Guangzhou, China
| | - Jin Chu
- MOE Key Laboratory of Laser Life Science, College of Biophotonics, South China Normal University, Guangzhou, China
| | - Xiaoyuan Deng
- MOE Key Laboratory of Laser Life Science, College of Biophotonics, South China Normal University, Guangzhou, China
| |
Collapse
|
33
|
Li Y, Wang X, Zhang L, Yuan X, Hao J, Ni J, Hao L. Upregulation of allograft inflammatory factor‑1 expression and secretion by macrophages stimulated with aldosterone promotes renal fibroblasts to a profibrotic phenotype. Int J Mol Med 2018; 42:861-872. [PMID: 29749461 PMCID: PMC6034929 DOI: 10.3892/ijmm.2018.3667] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2017] [Accepted: 05/04/2018] [Indexed: 02/07/2023] Open
Abstract
Macrophages have been identified as a key cell type in the pathogenesis of renal interstitial fibrosis (RIF). However, the mechanism through which macrophages drive fibrosis remains unclear. The current study focuses on the effects and possible underlying mechanism of allograft inflammatory factor-1 (AIF-1), an inflammation-responsive scaffold protein expressed and secreted by macrophages, in promoting fibroblasts to a profibrotic phenotype. In vivo experiments indicated that AIF-1, CD68 and α-smooth muscle actin (α-SMA) were upregulated in kidney tissues of mice subjected to unilateral ureteric obstruction, while their expressions were inhibited by an aldosterone receptor antagonist, spironolactone. Double immunofluorescence staining revealed that AIF-1 expression co-localized with CD68-positive macrophages in the renal interstitium, indicating that AIF-1 expression in macrophages was increased in the RIF animal model. Furthermore, to identify the role of AIF-1 in promoting fibrosis, its expression and secretion by the RAW264.7 macrophage cell line were detected in vitro. The expression levels of α-SMA, phosphorylated p38 (p-p38) and fibronectin (FN) in fibroblasts were examined subsequent to co-culture with macrophages. The increase in AIF-1 expression and secretion was confirmed in RAW264.7 cells in response to aldosterone. After 72 h of co-culture between fibroblasts and macrophages stimulated with aldosterone, the α-SMA expression was induced in fibroblasts, with significantly increased expression levels of FN and p-p38 observed. In addition, AIF-1 expression was reduced by stable transfection of RAW264.7 cells with AIF-1 small interfering RNA, resulting in significantly reduced expression levels of α-SMA, p-p38 and FN in fibroblasts co-cultured with macrophages as compared with normal macrophages. These findings indicate that the expression of AIF-1 in macrophages is critical for the activation of renal fibroblasts to a profibrotic phenotype. AIF-1 expression was upregulated in macrophages, and may be a novel mechanism linking macrophages to the promotion of RIF via the p38 signaling pathway.
Collapse
Affiliation(s)
- Yushu Li
- Department of Nephropathy, First Affiliated Hospital of Harbin Medical University, Harbin, Heilongjiang 150001, P.R. China
| | - Xingzhi Wang
- Department of Nephropathy, First Affiliated Hospital of Harbin Medical University, Harbin, Heilongjiang 150001, P.R. China
| | - Lei Zhang
- Department of Nephropathy, First Affiliated Hospital of Harbin Medical University, Harbin, Heilongjiang 150001, P.R. China
| | - Xueying Yuan
- Department of Nephropathy, First Affiliated Hospital of Harbin Medical University, Harbin, Heilongjiang 150001, P.R. China
| | - Jianbing Hao
- Department of Nephropathy, First Affiliated Hospital of Harbin Medical University, Harbin, Heilongjiang 150001, P.R. China
| | - Jie Ni
- Department of Nephropathy, First Affiliated Hospital of Harbin Medical University, Harbin, Heilongjiang 150001, P.R. China
| | - Lirong Hao
- Department of Nephropathy, First Affiliated Hospital of Harbin Medical University, Harbin, Heilongjiang 150001, P.R. China
| |
Collapse
|
34
|
Peiseler M, Kubes P. Macrophages play an essential role in trauma-induced sterile inflammation and tissue repair. Eur J Trauma Emerg Surg 2018; 44:335-349. [DOI: 10.1007/s00068-018-0956-1] [Citation(s) in RCA: 44] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2018] [Accepted: 04/12/2018] [Indexed: 12/20/2022]
|
35
|
Aberer E, Surtov-Pudar M, Wilfinger D, Deutsch A, Leitinger G, Schaider H. Co-culture of human fibroblasts and Borrelia burgdorferi enhances collagen and growth factor mRNA. Arch Dermatol Res 2018; 310:117-126. [PMID: 29214350 PMCID: PMC5811584 DOI: 10.1007/s00403-017-1797-1] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2017] [Revised: 11/02/2017] [Accepted: 11/17/2017] [Indexed: 11/18/2022]
Abstract
Skin fibrosis has been reported in Borrelia burgdorferi infection in Europe, but has been questioned by several authors. The objective of the present study was to examine the interaction of skin fibroblasts with B. burgdorferi sensu stricto B31 (BB) and B. afzelii (BA) in vitro by electron microscopy. We also determined the expression of collagen type I, TGF-β, FGF-1, calreticulin (CALR), decorin (DCN), and PDGF-α at the mRNA level in Borrelia/fibroblast co-cultures. Intact Borrelia attach to and transmigrate fibroblasts, and undergo cystic transformation outside the fibroblasts. Fibroblasts preserve their vitality and express a prominent granular endoplasmic reticulum, suggesting activated protein synthesis. On two different semi-quantitative real-time PCR assays, BB- and BA/fibroblast co-cultures showed a significant induction of type I collagen mRNA after 2 days compared to fibroblasts (fourfold for BA and 1.8-fold for BB; p < 0.02). In addition, there was a significant upregulation of mRNA expression of TGF-β, CALR, PDGF-α, and DCN in BA and BB co-cultures compared to control fibroblasts in monolayer cultures after 2 days (p < 0.01). The BA/fibroblast co-culture induced a considerably greater upregulation of collagen and growth factor mRNA compared to BB/fibroblast co-culture. In contrast, a significant down-regulation of FGF-1 (20-fold for BA and 4.5-fold for BB) mRNA expression was detected in co-cultures compared to controls (p < 0.01). The results of the study support the hypothesis that BB sensu lato, and BA in particular, enhances collagen mRNA expression and can stimulate growth factors responsible for increased collagen production.
Collapse
Affiliation(s)
- Elisabeth Aberer
- Department of Dermatology, Medical University of Graz, Auenbrugger Platz 8, 8036, Graz, Austria.
| | - Milana Surtov-Pudar
- Department of Dermatology, Medical University of Graz, Auenbrugger Platz 8, 8036, Graz, Austria
| | - Daniel Wilfinger
- Department of Dermatology, Medical University of Graz, Auenbrugger Platz 8, 8036, Graz, Austria
| | - Alexander Deutsch
- Division of Haematology, Internal Medicine, Medical University of Graz, Graz, Austria
| | - Gerd Leitinger
- Research Unit Electron-Microscopic Techniques, Institute of Cell Biology, Histology and Embryology, Medical University of Graz, Graz, Austria
| | - Helmut Schaider
- Department of Dermatology, Medical University of Graz, Auenbrugger Platz 8, 8036, Graz, Austria
- The University of Queensland Diamantina Institute, Translational Research Institute, University of Queensland, Brisbane, QLD, Australia
| |
Collapse
|
36
|
Clayton NS, Wilson AS, Laurent EP, Grose RP, Carter EP. Fibroblast growth factor-mediated crosstalk in cancer etiology and treatment. Dev Dyn 2017; 246:493-501. [PMID: 28470714 DOI: 10.1002/dvdy.24514] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2017] [Revised: 04/21/2017] [Accepted: 04/25/2017] [Indexed: 12/26/2022] Open
Abstract
It is becoming increasingly evident that multiple cell types within the tumor work together to drive tumour progression and impact on both the response to therapy and the dissemination of tumour cells throughout the body. Fibroblast growth factor signalling (FGF) is perturbed in a number of tumors, serving to drive tumor cell proliferation and migration, but also has a central role in orchestrating the plethora of cells that comprise the tumor microenvironment. This review focuses on how this family of signalling molecules can influence the interactions between tumor cells and their surrounding environment. Unraveling the complexities of FGF signalling between the distinct cell types of a tumor may identify additional opportunities for FGF-targeted compounds in therapy and could help combat drug resistance. Developmental Dynamics 246:493-501, 2017. © 2017 Wiley Periodicals, Inc.
Collapse
Affiliation(s)
- N S Clayton
- Centre for Tumour Biology, Barts Cancer Institute-a CRUK Centre of Excellence, Queen Mary University of London, London, United Kingdom
| | - A S Wilson
- Centre for Tumour Biology, Barts Cancer Institute-a CRUK Centre of Excellence, Queen Mary University of London, London, United Kingdom
| | - E P Laurent
- Centre for Tumour Biology, Barts Cancer Institute-a CRUK Centre of Excellence, Queen Mary University of London, London, United Kingdom
| | - R P Grose
- Centre for Tumour Biology, Barts Cancer Institute-a CRUK Centre of Excellence, Queen Mary University of London, London, United Kingdom
| | - E P Carter
- Centre for Tumour Biology, Barts Cancer Institute-a CRUK Centre of Excellence, Queen Mary University of London, London, United Kingdom
| |
Collapse
|
37
|
Abstract
Macrophages regulate tissue regeneration following injury. They can worsen tissue injury by producing reactive oxygen species and other toxic mediators that disrupt cell metabolism, induce apoptosis, and exacerbate ischemic injury. However, they also produce a variety of growth factors, such as IGF-1, VEGF-α, TGF-β, and Wnt proteins that regulate epithelial and endothelial cell proliferation, myofibroblast activation, stem and tissue progenitor cell differentiation, and angiogenesis. Proresolving macrophages in turn restore tissue homeostasis by functioning as anti-inflammatory cells, and macrophage-derived matrix metalloproteinases regulate fibrin and collagen turnover. However, dysregulated macrophage function impairs wound healing and contributes to the development of fibrosis. Consequently, the mechanisms that regulate these different macrophage activation states have become active areas of research. In this review, we discuss the common and unique mechanisms by which macrophages instruct tissue repair in the liver, nervous system, heart, lung, skeletal muscle, and intestine and illustrate how macrophages might be exploited therapeutically.
Collapse
Affiliation(s)
- Kevin M Vannella
- Immunopathogenesis Section, Laboratory of Parasitic Diseases, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, Maryland 20892; ,
| | - Thomas A Wynn
- Immunopathogenesis Section, Laboratory of Parasitic Diseases, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, Maryland 20892; ,
| |
Collapse
|
38
|
Skin fibrosis: Models and mechanisms. Curr Res Transl Med 2016; 64:185-193. [PMID: 27939457 DOI: 10.1016/j.retram.2016.06.003] [Citation(s) in RCA: 41] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2016] [Revised: 06/15/2016] [Accepted: 06/17/2016] [Indexed: 02/06/2023]
Abstract
Matrix synthesis, deposition and remodeling are complex biological processes that are critical in development, maintenance of tissue homeostasis and repair of injured tissues. Disturbances in the regulation of these processes can result in severe pathological conditions which are associated with tissue fibrosis as e.g. in Scleroderma, cutaneous Graft-versus-Host-Disease, excessive scarring after trauma or carcinogenesis. Therefore, finding efficient treatments to limit skin fibrosis is of major clinical importance. However the pathogenesis underlying the development of tissue fibrosis is still not entirely resolved. In recent years progress has been made unraveling the complex cellular and molecular mechanisms that determine fibrosis. Here we provide an overview of established and more recently developed mouse models that can be used to investigate the mechanisms of skin fibrosis and to test potential therapeutic approaches.
Collapse
|
39
|
Akahoshi K, Tanaka S, Mogushi K, Shimada S, Matsumura S, Akiyama Y, Aihara A, Mitsunori Y, Ban D, Ochiai T, Kudo A, Arii S, Tanabe M. Expression of connective tissue growth factor in the livers of non-viral hepatocellular carcinoma patients with metabolic risk factors. J Gastroenterol 2016; 51:910-22. [PMID: 26739296 DOI: 10.1007/s00535-015-1159-8] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/23/2015] [Accepted: 12/21/2015] [Indexed: 02/07/2023]
Abstract
BACKGROUND The incidence of hepatocellular carcinoma (HCC) associated with metabolic risk factors, such as diabetes and obesity, has been increasing. However, the underlying mechanism that links these diseases remains unclear. METHODS We performed genome-wide expression analysis of human liver tissues of non-viral HCC patients with or without metabolic risk factors. The upregulated genes that associated with diabetes and obesity were investigated by in vitro and in vivo experiments, and immunohistochemistry of human liver tissues was performed. RESULTS Among the upregulated genes, connective tissue growth factor (CTGF) expression was induced to a greater extent by combined glucose and insulin administration to human hepatoma cells. Genome-wide expression analysis revealed upregulation of a chemokine network in CTGF-overexpressing hepatoma cells, which displayed an increased ability to induce in vitro activation of macrophages, and in vivo infiltration of liver macrophages. Immunohistochemistry of human liver tissues validated the correlations between CTGF expression and diabetes or obesity as well as activation of liver macrophages in patients with non-viral HCC. Recurrence-free survival was significantly poorer in the CTGF-positive patients compared with the CTGF-negative patients (p = 0.002). Multivariate analysis determined that CTGF expression (HR 2.361; 95 % CI 1.195-4.665; p = 0.013) and vascular invasion (HR 2.367; 95 % CI 1.270-4.410; p = 0.007) were independent prognostic factors for recurrence of non-viral HCC. CONCLUSIONS Our data suggest that CTGF could be involved in oncogenic pathways promoting non-viral HCC associated with metabolic risk factors via induction of liver inflammation and is expected to be a novel HCC risk biomarker and potential therapeutic target.
Collapse
Affiliation(s)
- Keiichi Akahoshi
- Department of Molecular Oncology, Graduate School of Medicine, Tokyo Medical and Dental University, 1-5-45 Yushima, Bunkyo-ku, Tokyo, 113-8519, Japan.,Department of Hepato-Biliary-Pancreatic Surgery, Graduate School of Medicine, Tokyo Medical and Dental University, Tokyo, Japan
| | - Shinji Tanaka
- Department of Molecular Oncology, Graduate School of Medicine, Tokyo Medical and Dental University, 1-5-45 Yushima, Bunkyo-ku, Tokyo, 113-8519, Japan. .,Department of Hepato-Biliary-Pancreatic Surgery, Graduate School of Medicine, Tokyo Medical and Dental University, Tokyo, Japan.
| | - Kaoru Mogushi
- Department of Molecular Oncology, Graduate School of Medicine, Tokyo Medical and Dental University, 1-5-45 Yushima, Bunkyo-ku, Tokyo, 113-8519, Japan
| | - Shu Shimada
- Department of Molecular Oncology, Graduate School of Medicine, Tokyo Medical and Dental University, 1-5-45 Yushima, Bunkyo-ku, Tokyo, 113-8519, Japan
| | - Satoshi Matsumura
- Department of Hepato-Biliary-Pancreatic Surgery, Graduate School of Medicine, Tokyo Medical and Dental University, Tokyo, Japan
| | - Yoshimitsu Akiyama
- Department of Molecular Oncology, Graduate School of Medicine, Tokyo Medical and Dental University, 1-5-45 Yushima, Bunkyo-ku, Tokyo, 113-8519, Japan
| | - Arihiro Aihara
- Department of Hepato-Biliary-Pancreatic Surgery, Graduate School of Medicine, Tokyo Medical and Dental University, Tokyo, Japan
| | - Yusuke Mitsunori
- Department of Hepato-Biliary-Pancreatic Surgery, Graduate School of Medicine, Tokyo Medical and Dental University, Tokyo, Japan
| | - Daisuke Ban
- Department of Hepato-Biliary-Pancreatic Surgery, Graduate School of Medicine, Tokyo Medical and Dental University, Tokyo, Japan
| | - Takanori Ochiai
- Department of Hepato-Biliary-Pancreatic Surgery, Graduate School of Medicine, Tokyo Medical and Dental University, Tokyo, Japan
| | - Atsushi Kudo
- Department of Hepato-Biliary-Pancreatic Surgery, Graduate School of Medicine, Tokyo Medical and Dental University, Tokyo, Japan
| | - Shigeki Arii
- Department of Hepato-Biliary-Pancreatic Surgery, Graduate School of Medicine, Tokyo Medical and Dental University, Tokyo, Japan
| | - Minoru Tanabe
- Department of Hepato-Biliary-Pancreatic Surgery, Graduate School of Medicine, Tokyo Medical and Dental University, Tokyo, Japan
| |
Collapse
|
40
|
Huang YM, Chang PC, Wu SB, Kau HC, Tsai CC, Liu CJL, Wei YH. Expression and clinical significance of connective tissue growth factor (CTGF) in Graves' ophthalmopathy. Br J Ophthalmol 2016; 101:676-680. [PMID: 27543288 DOI: 10.1136/bjophthalmol-2016-308713] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2016] [Revised: 07/18/2016] [Accepted: 07/30/2016] [Indexed: 12/29/2022]
Abstract
AIMS To examine the expression of connective tissue growth factor (CTGF) in human cultured orbital fibroblasts from patients with Graves' ophthalmopathy (GO) and investigate whether a correlation exists between the presence of CTGF protein and clinical parameters of the disease. METHODS The protein expression levels of CTGF were analysed by western blots in cultured orbital fibroblasts from 10 patients with GO and 7 age-matched normal controls. Associations between the protein expression of CTGF and the clinical factors of GO, including clinical demographics, thyroid function, clinical activity score (CAS) and ophthalmopathy index (OI), was evaluated. RESULTS The mean protein expression levels of CTGF in the GO orbital fibroblasts were significantly higher than those of normal controls (p<0.001). Based on further analysis, the protein expression levels of CTGF in the GO orbital fibroblasts had significant correlation with gender (p=0.029), serum levels of thyrotropin receptor antibodies (p=0.029), CAS (p=0.048) and OI (p=0.043). Especially, there was a significant correlation between protein expression levels of CTGF and lid oedema (p=0.037), proptosis (p=0.045) and corneal involvement (p=0.001). CONCLUSIONS Our findings revealed that the protein expression levels of CTGF in the GO orbital fibroblasts were significantly highly expressed than those of normal controls, and the elevated CTGF was associated with clinical characteristics and evolution, indicating CTGF may play a role in the pathogenesis and pathophysiology of GO.
Collapse
Affiliation(s)
- Yi-Ming Huang
- Department of Ophthalmology, Taipei Veterans General Hospital and National Yang-Ming University, Taipei, Taiwan
| | - Pei-Chen Chang
- Institute of Clinical Medicine, National Yang-Ming University, Taipei, Taiwan
| | - Shi-Bei Wu
- Department of Biochemistry and Molecular Biology, National Yang-Ming University, Taipei, Taiwan
| | - Hui-Chuan Kau
- Department of Ophthalmology, Taipei Veterans General Hospital and National Yang-Ming University, Taipei, Taiwan.,Department of Ophthalmology, Koo Foundation Sun Yat-Sen Cancer Center, Taipei, Taiwan
| | - Chieh-Chih Tsai
- Department of Ophthalmology, Taipei Veterans General Hospital and National Yang-Ming University, Taipei, Taiwan
| | - Catherine Jui-Ling Liu
- Department of Ophthalmology, Taipei Veterans General Hospital and National Yang-Ming University, Taipei, Taiwan
| | - Yau-Huei Wei
- Institute of Clinical Medicine, National Yang-Ming University, Taipei, Taiwan.,Department of Medicine and Institute of Biomedical Sciences, Mackay Medical College, New Taipei City, Taiwan
| |
Collapse
|
41
|
Sakai S, Iwata C, Tanaka HY, Cabral H, Morishita Y, Miyazono K, Kano MR. Increased fibrosis and impaired intratumoral accumulation of macromolecules in a murine model of pancreatic cancer co-administered with FGF-2. J Control Release 2016; 230:109-15. [PMID: 27080571 DOI: 10.1016/j.jconrel.2016.04.007] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2015] [Revised: 02/29/2016] [Accepted: 04/05/2016] [Indexed: 02/07/2023]
Abstract
Pancreatic cancer is notorious for its poor prognosis. The histopathologic characteristic of pancreatic ductal adenocarcinoma (PDAC), which is the most common type of pancreatic cancer, is fibrosis within tumor tissue. Although fibrosis within tumor tissue is thought to impede drug therapy by interfering with the intratumoral accumulation of anti-tumor drugs, this hypothesis has yet to be proven directly in preclinical models. Here, we evaluated the effect of enhanced fibrosis on intratumoral accumulation of macromolecular drugs by increasing fibrosis in a murine tumor model of subcutaneously xenografted BxPC-3, a human PDAC cell line. When fibroblast growth factor-2 (FGF-2) was co-administered upon BxPC-3 inoculation, stromal fibrotic area was increased and was characterized by augmented murine collagen accumulation compared to inoculation of BxPC-3 alone, which correlated with increased monocyte/macrophage contents in the tumor tissues. We further discovered that the intratumoral accumulation of intravenously administrated fluorescein isothiocyanate-dextran of 2,000,000Da (2MDa) was significantly reduced in the FGF-2 co-administered tumors despite unaltered hyaluronan accumulation and pericyte coverage of the tumor neovasculature and increased lymphangiogenesis. Finally, we found that FGF-2 co-administered tumors are more refractory to macromolecular drug therapy using nab-paclitaxel (Abraxane). The model established and analyzed in this study, characterized by increased fibrotic component, provides a preclinical animal model suited to predict the intratumoral accumulation of macromolecular drugs and to evaluate the efficacy of drugs targeting the tumor stroma.
Collapse
Affiliation(s)
- Satoshi Sakai
- Department of Molecular Pathology, Graduate School of Medicine, The University of Tokyo, 7-3-1 Hongo, Bunkyo-ku, Tokyo 113-0033, Japan
| | - Caname Iwata
- Department of Molecular Pathology, Graduate School of Medicine, The University of Tokyo, 7-3-1 Hongo, Bunkyo-ku, Tokyo 113-0033, Japan
| | - Hiroyoshi Y Tanaka
- Department of Molecular Pathology, Graduate School of Medicine, The University of Tokyo, 7-3-1 Hongo, Bunkyo-ku, Tokyo 113-0033, Japan; Department of Pharmaceutical Biomedicine, Graduate School of Medicine, Dentistry, and Pharmaceutical Sciences, Okayama University, 1-1-1 Tsushima-Naka, Kita-ku, Okayama-shi, Okayama 700-8530, Japan
| | - Horacio Cabral
- Department of Bioengineering, Graduate School of Engineering, The University of Tokyo, 7-3-1 Hongo, Bunkyo-ku, Tokyo 113-0033, Japan
| | - Yasuyuki Morishita
- Department of Molecular Pathology, Graduate School of Medicine, The University of Tokyo, 7-3-1 Hongo, Bunkyo-ku, Tokyo 113-0033, Japan
| | - Kohei Miyazono
- Department of Molecular Pathology, Graduate School of Medicine, The University of Tokyo, 7-3-1 Hongo, Bunkyo-ku, Tokyo 113-0033, Japan
| | - Mitsunobu R Kano
- Department of Pharmaceutical Biomedicine, Graduate School of Medicine, Dentistry, and Pharmaceutical Sciences, Okayama University, 1-1-1 Tsushima-Naka, Kita-ku, Okayama-shi, Okayama 700-8530, Japan.
| |
Collapse
|
42
|
Wynn TA, Vannella KM. Macrophages in Tissue Repair, Regeneration, and Fibrosis. Immunity 2016; 44:450-462. [PMID: 26982353 PMCID: PMC4794754 DOI: 10.1016/j.immuni.2016.02.015] [Citation(s) in RCA: 2564] [Impact Index Per Article: 320.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2016] [Revised: 01/28/2016] [Accepted: 02/17/2016] [Indexed: 11/18/2022]
Abstract
Inflammatory monocytes and tissue-resident macrophages are key regulators of tissue repair, regeneration, and fibrosis. After tissue injury, monocytes and macrophages undergo marked phenotypic and functional changes to play critical roles during the initiation, maintenance, and resolution phases of tissue repair. Disturbances in macrophage function can lead to aberrant repair, such that uncontrolled production of inflammatory mediators and growth factors, deficient generation of anti-inflammatory macrophages, or failed communication between macrophages and epithelial cells, endothelial cells, fibroblasts, and stem or tissue progenitor cells all contribute to a state of persistent injury, and this could lead to the development of pathological fibrosis. In this review, we discuss the mechanisms that instruct macrophages to adopt pro-inflammatory, pro-wound-healing, pro-fibrotic, anti-inflammatory, anti-fibrotic, pro-resolving, and tissue-regenerating phenotypes after injury, and we highlight how some of these mechanisms and macrophage activation states could be exploited therapeutically.
Collapse
Affiliation(s)
- Thomas A Wynn
- Laboratory of Parasitic Diseases, National Institute of Allergy and Infectious Diseases, NIH, Bethesda, MD 20892, USA.
| | - Kevin M Vannella
- Laboratory of Parasitic Diseases, National Institute of Allergy and Infectious Diseases, NIH, Bethesda, MD 20892, USA
| |
Collapse
|
43
|
Miyashita T, Morimoto S, Fujishiro M, Hayakawa K, Suzuki S, Ikeda K, Miyazawa K, Morioka M, Takamori K, Ogawa H, Sekigawa I, Takasaki Y. Inhibition of each module of connective tissue growth factor as a potential therapeutic target for rheumatoid arthritis. Autoimmunity 2015; 49:109-14. [PMID: 26584911 DOI: 10.3109/08916934.2015.1113405] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Abstract
We previously reported the importance of connective tissue growth factor (CTGF) in rheumatoid arthritis (RA). CTGF contains four distinct modules connected in tandem, namely insulin-like growth factor-binding protein (IGFBP)-like, von Willebrand factor (vWF) type C repeat, thrombospondin type 1 (TSP-1) repeat, and carboxyl-terminal (CT) modules. The relationships between each of these modules of CTGF and RA remain unknown. Here, we analyzed how inhibition of each CTGF module affects the pathophysiology of RA. We conducted stimulation and suppression experiments on synovial cells (MH7A) obtained from patients with RA. Moreover, we examined angiogenesis by means of a tube-formation assay performed using human umbilical vein endothelial cells (HUVECs), and we used tartrate-resistant acid phosphatase (TRAP) staining to analyze osteoclastogenesis. Our results showed that M-CSF/RANKL-mediated osteoclastogenesis was enhanced when CTGF was added, but the effect of CTGF was neutralized by mAbs against CTGF modules 1-4. Furthermore, CTGF treatment of HUVECs induced formation of tubular networks, which resulted in acceleration of the angiogenesis of RA synoviocytes, and quantification showed that this tubular-network formation was also disrupted by anti-CTGF module 1-4 mAbs. Lastly, TNF-α enhanced the expression of CTGF and matrix metalloproteinase-3 (MMP3) in MH7A cells, and this enhancement was potently neutralized by mAbs against CTGF modules 1, 3 and 4. Thus, our results indicate that not only a mAb against CTGF but also mAbs against each specific module of CTGF might serve as potential therapeutic agents in the treatment of RA.
Collapse
Affiliation(s)
- Tomoko Miyashita
- a Institute for Environment and Gender Specific Medicine, Juntendo University Graduate School of Medicine , Chiba , Japan .,b Department of Internal Medicine and Rheumatology , School of Medicine, Juntendo University , Tokyo , Japan
| | - Shinji Morimoto
- a Institute for Environment and Gender Specific Medicine, Juntendo University Graduate School of Medicine , Chiba , Japan .,c Department of Internal Medicine and Rheumatology , Juntendo University Urayasu Hospital , Chiba , Japan
| | - Maki Fujishiro
- a Institute for Environment and Gender Specific Medicine, Juntendo University Graduate School of Medicine , Chiba , Japan
| | - Kunihiro Hayakawa
- a Institute for Environment and Gender Specific Medicine, Juntendo University Graduate School of Medicine , Chiba , Japan
| | - Satoshi Suzuki
- a Institute for Environment and Gender Specific Medicine, Juntendo University Graduate School of Medicine , Chiba , Japan .,b Department of Internal Medicine and Rheumatology , School of Medicine, Juntendo University , Tokyo , Japan
| | - Keigo Ikeda
- a Institute for Environment and Gender Specific Medicine, Juntendo University Graduate School of Medicine , Chiba , Japan .,c Department of Internal Medicine and Rheumatology , Juntendo University Urayasu Hospital , Chiba , Japan
| | - Keiji Miyazawa
- d Central Research Laboratories, Kissei Pharmaceutical Co Ltd , Nagano , Japan , and
| | | | - Kenji Takamori
- a Institute for Environment and Gender Specific Medicine, Juntendo University Graduate School of Medicine , Chiba , Japan
| | - Hideoki Ogawa
- a Institute for Environment and Gender Specific Medicine, Juntendo University Graduate School of Medicine , Chiba , Japan
| | - Iwao Sekigawa
- a Institute for Environment and Gender Specific Medicine, Juntendo University Graduate School of Medicine , Chiba , Japan .,c Department of Internal Medicine and Rheumatology , Juntendo University Urayasu Hospital , Chiba , Japan
| | - Yoshinari Takasaki
- b Department of Internal Medicine and Rheumatology , School of Medicine, Juntendo University , Tokyo , Japan
| |
Collapse
|
44
|
Ullah M, Cox S, Kelly E, Moore MAS, Zoellner H. Arecoline increases basic fibroblast growth factor but reduces expression of IL-1, IL-6, G-CSF and GM-CSF in human umbilical vein endothelium. J Oral Pathol Med 2014; 44:591-601. [PMID: 25529330 DOI: 10.1111/jop.12276] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 10/01/2014] [Indexed: 11/30/2022]
Abstract
BACKGROUND Areca nut chewing is associated with oral submucous fibrosis (OSF). Raised vascular basic fibroblast growth factor may induce fibrosis. Arecoline is a muscarinic alkaloid in areca nut, which we earlier reported causes injury and necrosis of human endothelium. MATERIALS AND METHODS Human umbilical vein endothelial cells were exposed to arecoline with or without tumor necrosis factor-α, and separately to acetylcholine, muscarine, or nicotine. Protein levels of basic fibroblast growth factor, as well as the inflammatory cytokines: granulocyte colony stimulating factor (G-CSF), granulocyte-macrophage colony stimulating factor, and Interleukins-6, 1-α and 1-β, were determined by enzyme-linked immunosorbent assay. mRNA levels were established by real-time reverse transcription polymerase chain reaction. RESULTS Basic fibroblast growth factor was released into the culture medium at arecoline levels causing necrosis (P < 0.05). This contrasted with an opposite effect of arecoline on levels of the inflammatory cytokines (P < 0.05). Tumor necrosis factor-α increased IL-6 and granulocyte-macrophage colony stimulated factor, but arecoline reduced this stimulated expression (P < 0.05). Arecoline had no effect on mRNA for basic fibroblast growth factor, although there was reduced mRNA for the separate inflammatory cytokines studied. The effect of acetylcholine, muscarine, and nicotine was minimal and dissimilar to that of arecoline. CONCLUSIONS Data raise the possibility that arecoline-induced, vascular basic fibroblast growth factor contributes to OSF, by combining increased growth factor expression with endothelial necrosis, and thus driving fibroblast proliferation.
Collapse
Affiliation(s)
- Mafaz Ullah
- The Cellular and Molecular Pathology Research Unit, The Faculty of Dentistry, Westmead Centre for Oral Health, Westmead Hospital, The University of Sydney, Westmead, NSW, Australia
| | - Stephen Cox
- The Department of Oral Surgery, The Faculty of Dentistry, Westmead Centre for Oral Health, Westmead Hospital, The University of Sydney, Westmead, NSW, Australia
| | - Elizabeth Kelly
- The Cellular and Molecular Pathology Research Unit, The Faculty of Dentistry, Westmead Centre for Oral Health, Westmead Hospital, The University of Sydney, Westmead, NSW, Australia
| | | | - Hans Zoellner
- The Cellular and Molecular Pathology Research Unit, The Faculty of Dentistry, Westmead Centre for Oral Health, Westmead Hospital, The University of Sydney, Westmead, NSW, Australia.,The Memorial Sloan Kettering Cancer Center, New York, NY, USA
| |
Collapse
|
45
|
Ehashi T, Kakinoki S, Yamaoka T. Water absorbing and quick degradable PLLA/PEG multiblock copolymers reduce the encapsulation and inflammatory cytokine production. J Artif Organs 2014; 17:321-8. [DOI: 10.1007/s10047-014-0791-z] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2014] [Accepted: 08/22/2014] [Indexed: 12/22/2022]
|
46
|
Abstract
Without doubt, animal models have provided significant insights into our understanding of the rheumatological diseases; however, no model has accurately replicated all aspects of any autoimmune disease. Recent years have seen a plethora of knockouts and transgenics that have contributed to our knowledge of the initiating events of systemic sclerosis, an autoimmune disease. In this review, the focus is on models of systemic sclerosis and how they have progressed our understanding of fibrosis and vasculopathy, and whether they are relevant to the pathogenesis of systemic sclerosis.
Collapse
Affiliation(s)
- Carol M Artlett
- Department of Microbiology and Immunology, Drexel University College of Medicine, Philadelphia, PA, USA
| |
Collapse
|
47
|
Matsushita T, Fujimoto M. Scleroderma: recent lessons from murine models and implications for future therapeutics. ACTA ACUST UNITED AC 2014. [DOI: 10.1586/17469872.2013.835924] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/06/2023]
|
48
|
Ho YY, Baron M, Recklies AD, Roughley PJ, Mort JS. Cells from the skin of patients with systemic sclerosis secrete chitinase 3-like protein 1. BBA CLINICAL 2014; 1:2-11. [PMID: 26675476 PMCID: PMC4633946 DOI: 10.1016/j.bbacli.2013.12.001] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 11/04/2013] [Revised: 12/18/2013] [Accepted: 12/19/2013] [Indexed: 12/22/2022]
Abstract
Background The chitinase-like protein, Chi3L1, is associated with increased fibrotic activity as well as inflammatory processes. The capacity of skin cells from systemic sclerosis (SSc) patients to produce Chi3L1, and the stimulation of its synthesis by cytokines or growth factors known to be associated with SSc, was investigated. Methods Cells were isolated from forearm and/or abdomen skin biopsies taken from SSc patients and normal individuals and stimulated with cytokines and growth factors to assess Chi3L1 expression. Chi3L1-expressing cells were characterized by immunohistochemical staining. Results Chi3L1 was not secreted by skin cells from normal individuals nor was its synthesis induced by any of the cytokines or growth factors investigated. In contrast, Chi3L1 secretion was induced by OSM or IL-1 in cells from all forearm biopsies of SSc patients, and endogenous secretion in the absence of cytokines was detected in several specimens. Patients with Chi3L1-producing cells at both the arm and abdomen had a disease duration of less than 3 years. Endogenous Chi3L1 production was not a property of the major fibroblast population nor of myofibroblasts, but rather was related to the presence of stem-like cells not present in normal skin. Other cells, however, contributed to the upregulation of Chi3L1 by OSM. Conclusions The emergence of cells primed to respond to OSM with increased Chi3L1 production appears to be associated with pathological processes active in SSc. General significance The presence of progenitor cells expressing the chilectin Chi3L1 in SSc skin appears to play a role in the initiation of the disease process. Cells isolated from the skin of scleroderma patients secrete Chi3L1. Chi3L1 production is stimulated by oncostatin M or interleukin 1. Patients with Chi3L1 producing cells have disease duration of < 3 years. Chi3L1 production is a property of stem-like cells not present in normal skin. Other cells contribute to Chi3L1 upregulation by oncostatin M.
Collapse
Key Words
- Chi3L1, chitinase 3-like protein 1
- Chitinase 3-like protein 1
- Cytokine
- DAPI, 4′,6-diamidino-2-phenylindole
- ECM, extracellular matrix
- IL, interleukin
- OSM, oncostatin M
- Oncostatin M
- PDGF, platelet-derived growth factor
- SBTI, soybean trypsin inhibitor
- SSc, systemic sclerosis (scleroderma)
- Scleroderma
- Stem cell
- Systemic sclerosis
- TGFβ, transforming growth factor-β
- TIE2, tyrosine kinase with Ig and EGF homology domains-2
- mRSS, modified Rodnan skin score
- αSMA, α-smooth muscle actin
Collapse
Affiliation(s)
- Yuen Yee Ho
- Shriners Hospital for Children, Department of Surgery, McGill University, 1529 Cedar Avenue, Montréal, Quebec H3G 1A6, Canada
| | - Murray Baron
- Department of Rheumatology, Jewish General Hospital, 3755 Cote Ste Catherine Road, Montréal, Quebec H3T 1E2, Canada
| | - Anneliese D Recklies
- Shriners Hospital for Children, Department of Surgery, McGill University, 1529 Cedar Avenue, Montréal, Quebec H3G 1A6, Canada
| | - Peter J Roughley
- Shriners Hospital for Children, Department of Surgery, McGill University, 1529 Cedar Avenue, Montréal, Quebec H3G 1A6, Canada
| | - John S Mort
- Shriners Hospital for Children, Department of Surgery, McGill University, 1529 Cedar Avenue, Montréal, Quebec H3G 1A6, Canada
| |
Collapse
|
49
|
Nozawa K, Fujishiro M, Kawasaki M, Yamaguchi A, Ikeda K, Morimoto S, Iwabuchi K, Yanagida M, Ichinose S, Morioka M, Ogawa H, Takamori K, Takasaki Y, Sekigawa I. Inhibition of Connective Tissue Growth Factor Ameliorates Disease in a Murine Model of Rheumatoid Arthritis. ACTA ACUST UNITED AC 2013; 65:1477-86. [DOI: 10.1002/art.37902] [Citation(s) in RCA: 32] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2012] [Accepted: 02/07/2013] [Indexed: 11/05/2022]
Affiliation(s)
- Kazuhisa Nozawa
- Juntendo University School of Medicine, Tokyo, Japan, and Juntendo University Graduate School of Medicine; Chiba; Japan
| | - Maki Fujishiro
- Juntendo University Graduate School of Medicine; Chiba; Japan
| | - Mikiko Kawasaki
- Juntendo University Graduate School of Medicine; Chiba; Japan
| | - Ayako Yamaguchi
- Juntendo University School of Medicine, Tokyo, Japan, and Juntendo University Graduate School of Medicine; Chiba; Japan
| | - Keigo Ikeda
- Juntendo University Graduate School of Medicine and Juntendo University Urayasu Hospital; Chiba; Japan
| | - Shinji Morimoto
- Juntendo University Graduate School of Medicine and Juntendo University Urayasu Hospital; Chiba; Japan
| | - Kazuhisa Iwabuchi
- Juntendo University School of Medicine, Tokyo, Japan, and Juntendo University Graduate School of Medicine; Chiba; Japan
| | | | - Shouzo Ichinose
- Juntendo University Graduate School of Medicine; Chiba; Japan
| | | | - Hideoki Ogawa
- Juntendo University Graduate School of Medicine; Chiba; Japan
| | - Kenji Takamori
- Juntendo University Graduate School of Medicine; Chiba; Japan
| | | | - Iwao Sekigawa
- Juntendo University Graduate School of Medicine and Juntendo University Urayasu Hospital; Chiba; Japan
| |
Collapse
|
50
|
Finger EC, Cheng CF, Williams TR, Rankin EB, Bedogni B, Tachiki L, Spong S, Giaccia AJ, Powell MB. CTGF is a therapeutic target for metastatic melanoma. Oncogene 2013; 33:1093-100. [PMID: 23435419 DOI: 10.1038/onc.2013.47] [Citation(s) in RCA: 55] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2012] [Revised: 12/19/2012] [Accepted: 01/11/2013] [Indexed: 02/07/2023]
Abstract
Metastatic melanoma remains a devastating disease with a 5-year survival rate of less than five percent. Despite recent advances in targeted therapies for melanoma, only a small percentage of melanoma patients experience durable remissions. Therefore, it is critical to identify new therapies for the treatment of advanced melanoma. Here, we define connective tissue growth factor (CTGF) as a therapeutic target for metastatic melanoma. Clinically, CTGF expression correlates with tumor progression and is strongly induced by hypoxia through HIF-1 and HIF-2-dependent mechanisms. Genetic inhibition of CTGF in human melanoma cells is sufficient to significantly reduce orthotopic tumor growth, as well as metastatic tumor growth in the lung of severe combined immunodeficient (SCID) mice. Mechanistically, inhibition of CTGF decreased invasion and migration associated with reduced matrix metalloproteinase-9 expression. Most importantly, the anti-CTGF antibody, FG-3019, had a profound inhibitory effect on the progression of established metastatic melanoma. These results offer the first preclinical validation of anti-CTGF therapy for the treatment of advanced melanoma and underscore the importance of tumor hypoxia in melanoma progression.
Collapse
Affiliation(s)
- E C Finger
- Division of Radiation and Cancer Biology, Department of Radiation Oncology, Stanford University, Stanford, CA, USA
| | - C-F Cheng
- Division of Radiation and Cancer Biology, Department of Radiation Oncology, Stanford University, Stanford, CA, USA
| | - T R Williams
- Division of Radiation and Cancer Biology, Department of Radiation Oncology, Stanford University, Stanford, CA, USA
| | - E B Rankin
- Division of Radiation and Cancer Biology, Department of Radiation Oncology, Stanford University, Stanford, CA, USA
| | - B Bedogni
- Department of Biochemistry, Case Western Reserve University School of Medicine, Cleveland, OH, USA
| | - L Tachiki
- Division of Radiation and Cancer Biology, Department of Radiation Oncology, Stanford University, Stanford, CA, USA
| | - S Spong
- FibroGen Inc., San Francisco, CA, USA
| | - A J Giaccia
- Division of Radiation and Cancer Biology, Department of Radiation Oncology, Stanford University, Stanford, CA, USA
| | - M B Powell
- Division of Radiation and Cancer Biology, Department of Radiation Oncology, Stanford University, Stanford, CA, USA
| |
Collapse
|