1
|
Sanpinit S, Issuriya A, Sakulkeo O, Wetchakul P, Limsuwan S, Na-Phatthalung P, Kantisin S, Tang J, Chusri S. Mechanisms underlying the wound healing and tissue regeneration properties of a novel gauze dressing impregnated with traditional herbal medicine (Ya-Samarn-Phlae) in type 2 diabetic Goto-Kakizaki (GK) rats. Front Pharmacol 2025; 16:1574715. [PMID: 40271074 PMCID: PMC12015241 DOI: 10.3389/fphar.2025.1574715] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2025] [Accepted: 04/01/2025] [Indexed: 04/25/2025] Open
Abstract
Ethnopharmacological relevance A traditional preparation of Ya-Samarn-Phlae (T-YaSP) consists of Garcinia mangostana L., Oryza sativa L., Curcuma longa L., and Areca catechu L. and has been used in Thai medicine as an infused oil for treating chronic and diabetic wounds. It is reputed for its antibacterial, antioxidant, and wound-healing properties. Despite its traditional use, scientific validation of the mechanisms underlying diabetic wound healing remains limited. Aim This study aims to develop a novel gauze dressing impregnated with an ointment containing T-YaSP (YaSP) to enhance its practical application and elucidate the mechanisms of action in promoting wound healing in both non-diabetic and type 2 diabetic wounds of this ointment. Materials and methods YaSP was developed and tested for stability and dermal irritation. Changes in chemical markers during storage were measured both qualitatively and quantitatively. Its anti-inflammatory activity was assessed using the carrageenan-induced rat paw edema model. The effect of YaSP on levels of nitric oxide (NO), myeloperoxidase (MPO), malondialdehyde (MDA), inflammatory cytokines (TNF-α, IL-1β, and PGE2), and pro-inflammatory enzymes (iNOS and COX-2) was measured. The wound-healing effects of YaSP were assessed using full-thickness (6 mm diameter) wound models in both non-diabetic Wistar rats and type 2 diabetic Goto-Kakizaki rats. In addition to evaluating wound closure on days 0, 3, 5, 7, 9, and 11, the influence on TGF-β1, VEGF, and the production of collagen types I and III, which indicate the inflammatory, proliferative, and remodeling phases, was measured. Results During the 6-month storage period, the α-mangostin content measured in YaSP did not decrease; however, the curcumin level showed a significant reduction. Topical treatment with YaSP demonstrated strong anti-inflammatory activity and alleviated oxidative stress and inflammatory markers. YaSP improved wound closure rates in both diabetic and non-diabetic models. Levels of TGF-β1 and VEGF increased, indicating the promotion of angiogenesis and granulation tissue formation during the proliferation phase on the seventh day. Additionally, TGF-β1 levels dropped on the 11th day, aligning with diminished inflammation and enhanced remodeling. The treatment balanced collagen synthesis, increasing type III collagen in the early stages and type I collagen in the later stages of wound healing. Histological analysis confirmed reduced inflammation, enhanced neovascularization, and increased collagen production. Conclusion A gauze dressing impregnated with YaSP provides a practical solution for diabetic wound management and demonstrates strong wound-healing properties by modulating excess inflammation, promoting angiogenesis during the proliferation phase, and regulating collagen synthesis throughout the remodeling phase. This discovery reveals, for the first time, the underlying mechanisms of action of this traditional formulation, highlighting its potential as a cost-effective alternative for managing chronic wounds in resource-limited settings.
Collapse
Affiliation(s)
- Sineenart Sanpinit
- Department of Applied Thai Traditional Medicine, School of Medicine, Walailak University, Thasala, Nakhon Si Thammarat, Thailand
| | - Acharaporn Issuriya
- Division of Health and Applied Sciences, Faculty of Science, Prince of Songkla University, Songkhla, Thailand
| | - Oraphan Sakulkeo
- Traditional Thai Medical Research and Innovation Center, Faculty of Traditional Thai Medicine, Prince of Songkla University, Hat Yai, Songkhla, Thailand
| | - Palika Wetchakul
- Department of Applied Thai Traditional Medicine, School of Medicine, Walailak University, Thasala, Nakhon Si Thammarat, Thailand
| | - Surasak Limsuwan
- Traditional Thai Medical Research and Innovation Center, Faculty of Traditional Thai Medicine, Prince of Songkla University, Hat Yai, Songkhla, Thailand
| | - Pinanong Na-Phatthalung
- Division of Hematology and Oncology, Icahn School of Medicine at Mount Sinai, New York, NY, United States
| | - Siriwan Kantisin
- Unit for Area-Based Research and Innovation in Cross-Border Health Care and Occupational Health and Safety Department, School of Health Science, Mae Fah Luang University, Chiang Rai, Thailand
| | - Jian Tang
- School of Chinese Medicine, Bozhou University, Bozhou, China
| | - Sasitorn Chusri
- School of Health Science and Biomedical Technology Research Group for Vulnerable Populations, Mae Fah Luang University, Chiang Rai, Thailand
| |
Collapse
|
2
|
Williams R. COVID-19 Humic/Fulvic Acid Plus Epigallocatechin Gallate Treatment: A Retrospective Chart Review. Cureus 2025; 17:e77188. [PMID: 39925527 PMCID: PMC11806964 DOI: 10.7759/cureus.77188] [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: 10/16/2024] [Accepted: 01/09/2025] [Indexed: 02/11/2025] Open
Abstract
INTRODUCTION The initial COVID-19 infection caused significant mortality in the nursing home setting. The mortality in our facility was approximately 50% and occurred in the later phase of the initial infection. In the nursing home setting, there were no approved treatment regimes. Specific treatment was left to the treating physician in concert with the administrative medical officials; immunizations were approved in December 2020. Concurrent publications indicated that epigallocatechin gallate (EGCG) and humic/fulvic acid with vitamin C possess the ability to interfere with the SARS-CoV-2 virus; both can inhibit viral attachment and reproduction, along with providing significant anti-inflammatory activity. They also exhibit good safety profiles and were previously utilized in our facility. With the high mortality and significant published data on inhibitory aspects of EGCG, humic/fulvic acid, and vitamin C, our facility utilized these supplements to treat COVID-19-positive patients. METHODS This retrospective chart review analyzes the effectiveness of an integrative treatment consisting of EGCG and humic/fulvic acid with vitamin C provided to COVID-19 patients in a long-term care facility between October and December 2020. Nursing home patients with COVID-19 infections were either provided integrative treatment utilizing EGCG, humic/fulvic acid, and vitamin C or provided care without integrative support. All EGCG patients were informed, and, when needed, the first line of authority, power of attorney (POA) was contacted and informed that the integrative approach was not standard treatment (but was reviewed and approved by Pulaski Health and Rehabilitation Medical Facility directors and the patient's physician). Patients were informed of the safety literature and compounds' utilization in viral infections prior to accepting treatment. RESULTS A review of 60 records indicates that among 22 patients receiving treatment with the integrative combination, there were no mortalities. Among 38 patients treated without this integrative support, there were 21 deaths. Conclusions: In a retrospective review of the treatment data, treatment of COVID-19 in the nursing home setting with EGCG, humic/fulvic acid, and vitamin C resulted in a significant reduction in overall mortality.
Collapse
Affiliation(s)
- Richard Williams
- Geriatrics, Edward Via College of Osteopathic Medicine, Blacksburg, USA
| |
Collapse
|
3
|
Zhang Y, Liu E, Gao H, He Q, Chen A, Pang Y, Zhang X, Bai S, Zeng J, Guo J. Natural products for the treatment of hypertrophic scars: Preclinical and clinical studies. Heliyon 2024; 10:e37059. [PMID: 39296083 PMCID: PMC11408005 DOI: 10.1016/j.heliyon.2024.e37059] [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: 03/12/2024] [Revised: 08/13/2024] [Accepted: 08/27/2024] [Indexed: 09/21/2024] Open
Abstract
Hypertrophic scarring (HS) is a complication of wound healing that causes physiological and psychological distress in patients. However, the possible mechanism underlying HS is not fully understood, and there is no gold standard for its treatment. Natural products are more effective, economical, convenient, and safe than existing drugs, and they have a wide application prospect. However, there is a lack of literature on this topic, so we reviewed in vivo, in vitro, and clinical studies and screened natural products showing beneficial effects on HS that can become potential therapeutic agents for HS to fill in the gaps in the field. In addition, we discussed the drug delivery systems related to these natural products and their mechanisms in the treatment of HS. Generally speaking, natural products inhibit inflammation, myofibroblast activation, angiogenesis, and collagen accumulation by targeting interleukins, tumor necrosis factor-α, vascular endothelial growth factors, platelet-derived growth factors, and matrix metalloproteinases, so as to play an anti-HS effects of natural products are attributed to their anti-inflammatory, anti-proliferative, anti-angiogenesis, and pro-apoptotic (enhancing apoptosis and autophagy) roles, thus treating HS. We also screened the potential therapeutic targets of these natural compounds for HS through network pharmacology and constructed a protein-protein interaction (PPI) network, which may provide clues for the pharmacological mechanism of natural products in treating this disease and the development and application of drugs.
Collapse
Affiliation(s)
- Yuxiao Zhang
- Hospital of Chengdu University of Traditional Chinese Medicine Department of Dermatology, China
| | - E Liu
- Hospital of Chengdu University of Traditional Chinese Medicine Department of Dermatology, China
| | | | - Qingying He
- Hospital of Chengdu University of Traditional Chinese Medicine Department of Dermatology, China
| | - Anjing Chen
- Hospital of Chengdu University of Traditional Chinese Medicine Department of Dermatology, China
| | - Yaobing Pang
- Hospital of Chengdu University of Traditional Chinese Medicine Department of Dermatology, China
| | - Xueer Zhang
- Hospital of Chengdu University of Traditional Chinese Medicine Department of Dermatology, China
| | - Sixian Bai
- Hospital of Chengdu University of Traditional Chinese Medicine Department of Dermatology, China
| | - Jinhao Zeng
- Hospital of Chengdu University of Traditional Chinese Medicine Department of Dermatology, China
| | - Jing Guo
- Hospital of Chengdu University of Traditional Chinese Medicine Department of Dermatology, China
| |
Collapse
|
4
|
Evangelisti G, Ferrero S, Perrone U, Gustavino C, Volpi E, Izzotti A, Barra F. Experimental and new investigational drugs for the treatment of uterine fibroids. Expert Opin Investig Drugs 2024; 33:497-508. [PMID: 38618931 DOI: 10.1080/13543784.2024.2343786] [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: 12/09/2023] [Accepted: 04/12/2024] [Indexed: 04/16/2024]
Abstract
INTRODUCTION Uterine fibroids, the most prevalent benign tumors among reproductive-age women, pose treatment challenges that range from surgical interventions to medical therapies for symptom control. Progestins and estroprogestins effectively manage uterine bleeding by suppressing dysfunctional endometrium over fibroids. While GnRH agonists represent a crucial milestone in symptom treatment, their prolonged use results in menopausal-like symptoms and irreversible bone mineral density loss. Advancements in understanding fibroid pathophysiology have prompted the exploration of new compounds to overcome current therapy limitations. AREAS COVERED This manuscript offers an updated overview of investigational drugs for symptomatic uterine fibroids. EXPERT OPINION Despite ulipristal acetate's well-established efficacy as a selective progesterone receptor modulator (SPRM) in fibroid treatment, its prescription has declined due to the rare but severe risk of liver damage. Oral GnRH antagonists, like elagolix, relugolix, and linzagolix, with their novel pharmacodynamic properties, are gaining traction in fibroid management, inducing a dose-dependent reduction in circulating sex hormone levels. Ongoing research on natural compounds, such as vitamin D and epigallocatechin gallate (EGCG), presents emerging options for treating uterine fibroids. This evolving landscape reflects the ongoing efforts to improve therapeutic outcomes for individuals with symptomatic uterine fibroids.
Collapse
Affiliation(s)
- Giulio Evangelisti
- Unit of Obstetrics and Gynecology, San Paolo Hospital - ASL2, Savona, Italy
- Department of Neurosciences, Rehabilitation, Ophthalmology, Genetics, Maternal and Child Health (DiNOGMI), University of Genoa, Genoa, Italy
| | - Simone Ferrero
- Department of Neurosciences, Rehabilitation, Ophthalmology, Genetics, Maternal and Child Health (DiNOGMI), University of Genoa, Genoa, Italy
- Academic Unit of Obstetrics and Gynecology, IRCCS Ospedale Policlinico San Martino, Genoa, Italy
| | - Umberto Perrone
- Department of Neurosciences, Rehabilitation, Ophthalmology, Genetics, Maternal and Child Health (DiNOGMI), University of Genoa, Genoa, Italy
- Academic Unit of Obstetrics and Gynecology, IRCCS Ospedale Policlinico San Martino, Genoa, Italy
| | - Claudio Gustavino
- Unit of Obstetrics and Gynecology, IRCCS Ospedale Policlinico San Martino, Genoa, Italy
| | - Eugenio Volpi
- Unit of Obstetrics and Gynecology, San Paolo Hospital - ASL2, Savona, Italy
| | - Alberto Izzotti
- Unit of Mutagenesis and Cancer Prevention, IRCCS Ospedale Policlinico San Martino, Genoa, Italy
- Department of Experimental Medicine (DIMES), University of Genoa, Genoa, Italy
| | - Fabio Barra
- Unit of Obstetrics and Gynecology, P.O. "Ospedale del Tigullio"- ASL4, Chiavari, Genoa, Italy
- Department of Health Sciences (DISSAL), University of Genoa, Genoa, Italy
| |
Collapse
|
5
|
Abstract
Fibrosis is a common and debilitating pathological process that affects many organ systems and contributes to connective tissue disorders in orthopaedics. Tendons heal after acute and chronic injury through a process of fibrovascular scar tissue formation, and soft tissue joint capsules can be affected after traumatic joint injury, leading to arthrofibrosis. Although the precise underlying mechanisms are still being elucidated, fibrosis is thought to be a consequence of dysregulated immune and cytokine signaling that leads to myofibroblast activation and proliferation and subsequent excessive collagen deposition. Current treatments for connective tissue fibrosis include physical therapy and surgery, but there are no therapies that directly target the underlying cellular and molecular mechanisms of fibrosis. Many pharmacological agents have been used to successfully target fibrosis in other tissues and organ systems and thus are a promising treatment option to fill this gap. However, limited evidence is available to guide the use of these agents in musculoskeletal connective tissues. This article provides an overview of pharmacological therapies that have potential to treat connective tissue fibrosis in patients with musculoskeletal conditions, along with the current supporting evidence and future uses of each therapy.
Collapse
Affiliation(s)
- Nathaniel P Disser
- Hospital for Special Surgery, New York, New York, USA
- McGovern Medical School, The University of Texas Health Science Center at Houston, Houston, Texas, USA
| | - Jonathan S Yu
- Hospital for Special Surgery, New York, New York, USA
- Weill Cornell Medicine, New York, New York, USA
| | - Vincent J H Yao
- Hospital for Special Surgery, New York, New York, USA
- Sophie Davis Biomedical Education Program at CUNY School of Medicine, New York, New York, USA
| | - Scott A Rodeo
- Hospital for Special Surgery, New York, New York, USA
| |
Collapse
|
6
|
Wang MC. Natural plant resource flavonoids as potential therapeutic drugs for pulmonary fibrosis. Heliyon 2023; 9:e19308. [PMID: 37664726 PMCID: PMC10470008 DOI: 10.1016/j.heliyon.2023.e19308] [Citation(s) in RCA: 12] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2023] [Revised: 08/04/2023] [Accepted: 08/17/2023] [Indexed: 09/05/2023] Open
Abstract
Pulmonary fibrosis is an enduring and advancing pulmonary interstitial disease caused by multiple factors that ultimately lead to structural changes in normal lung tissue. Currently, pulmonary fibrosis is a global disease with a high degree of heterogeneity and mortality rate. Nitidine and pirfenidone have been approved for treating pulmonary fibrosis, and the quest for effective therapeutic drugs remains unabated. In recent years, the anti-pulmonary fibrosis properties of natural flavonoids have garnered heightened attention, although further research is needed. In this paper, the resources, structural characteristics, anti-pulmonary fibrosis properties and mechanisms of natural flavonoids were reviewed. We hope to provide potential opportunities for the application of flavonoids in the fight against pulmonary fibrosis.
Collapse
Affiliation(s)
- Meng-Chuan Wang
- Department of Pharmacy, Affiliated Cixi Hospital, Wenzhou Medical University, China
| |
Collapse
|
7
|
Hazimeh D, Massoud G, Parish M, Singh B, Segars J, Islam MS. Green Tea and Benign Gynecologic Disorders: A New Trick for An Old Beverage? Nutrients 2023; 15:1439. [PMID: 36986169 PMCID: PMC10054707 DOI: 10.3390/nu15061439] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2023] [Revised: 03/08/2023] [Accepted: 03/14/2023] [Indexed: 03/19/2023] Open
Abstract
Green tea is harvested from the tea plant Camellia sinensis and is one of the most widely consumed beverages worldwide. It is richer in antioxidants than other forms of tea and has a uniquely high content of polyphenolic compounds known as catechins. Epigallocatechin-3-gallate (EGCG), the major green tea catechin, has been studied for its potential therapeutic role in many disease contexts, including pathologies of the female reproductive system. As both a prooxidant and antioxidant, EGCG can modulate many cellular pathways important to disease pathogenesis and thus has clinical benefits. This review provides a synopsis of the current knowledge on the beneficial effects of green tea in benign gynecological disorders. Green tea alleviates symptom severity in uterine fibroids and improves endometriosis through anti-fibrotic, anti-angiogenic, and pro-apoptotic mechanisms. Additionally, it can reduce uterine contractility and improve the generalized hyperalgesia associated with dysmenorrhea and adenomyosis. Although its role in infertility is controversial, EGCG can be used as a symptomatic treatment for menopause, where it decreases weight gain and osteoporosis, as well as for polycystic ovary syndrome (PCOS).
Collapse
Affiliation(s)
| | | | | | | | - James Segars
- Department of Gynecology and Obstetrics, Division of Reproductive Sciences & Women’s Health Research, Johns Hopkins Medicine, Baltimore, MD 21205, USA
| | - Md Soriful Islam
- Department of Gynecology and Obstetrics, Division of Reproductive Sciences & Women’s Health Research, Johns Hopkins Medicine, Baltimore, MD 21205, USA
| |
Collapse
|
8
|
Dinda B, Dinda S, Dinda M. Therapeutic potential of green tea catechin, (-)-epigallocatechin-3- O-gallate (EGCG) in SARS-CoV-2 infection: Major interactions with host/virus proteases. PHYTOMEDICINE PLUS : INTERNATIONAL JOURNAL OF PHYTOTHERAPY AND PHYTOPHARMACOLOGY 2023; 3:100402. [PMID: 36597465 PMCID: PMC9800022 DOI: 10.1016/j.phyplu.2022.100402] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 12/12/2022] [Accepted: 12/24/2022] [Indexed: 06/17/2023]
Abstract
BACKGROUND The current COVID-19 pandemic from the human pathogenic virus SARS-CoV-2 has resulted in a major health hazard globally. The morbidity and transmission modality of this disease are severe and uncontrollable. As no effective clinical drugs are available for treatment of COVID-19 infection till to date and only vaccination is used as prophylaxis and its efficacy is restricted due to emergent of new variants of SARS-CoV-2, there is an urgent need for effective drugs for its treatment. PURPOSE The aim of this review was to provide a detailed analysis of anti-SARS-CoV-2 efficacy of (-)-epigallocatechin-3-O-gallate (EGCG), a major catechin constituent of green tea (Camellia sinensis (L.) Kuntze) beverage to highlight the scope of EGCG in clinical medicine as both prophylaxis and treatment of present COVID-19 infection. In addition, the factors related to poor oral bioavailabilty of EGCG was also analysed for a suggestion for future research in this direction. STUDY DESIGN We collected the published articles related to anti-SARS-CoV-2 activity of EGCG against the original strain (Wuhan type) and its newly emerged variants of SARS-CoV-2 virus. METHODS A systematic search on the published literature was conducted in various databases including Google Scholar, PubMed, Science Direct and Scopus to collect the relevant literature. RESULTS The findings of this search demonstrate that EGCG shows potent antiviral activity against SARS-CoV-2 virus by preventing viral entry and replication in host cells in vitro models. The studies on the molecular mechanisms of EGCG in inhibition of SARS-CoV-2 infection in host cells reveal that EGCG blocks the entry of the virus particles by interaction with the receptor binding domain (RBD) of viral spike (S) protein to host cell surface receptor protease angiotensin-converting enzyme 2 (ACE2) as well as suppression of the expressions of host proteases, ACE2, TMPRSS2 and GRP78, required for viral entry, by Nrf2 activation in host cells. Moreover, EGCG inhibits the activities of SARS-CoV-2 main protease (Mpro), papain-like protease (PLpro), endoribonuclease Nsp15 in vitro models and of RNA-dependent RNA polymerase (RdRp) in molecular docking model for suppression of viral replication. In addition, EGCG significantly inhibits viral inflammatory cytokine production by stimulating Nrf2- dependent host immune response in virus-infected cells. EGCG significantly reduces the elevated levels of HMGB1, a biomarker of sepsis, lung fibrosis and thrombotic complications in viral infections. EGCG potentially inhibits the infection of original (Wuhan type) strain of SARS-CoV-2 and other newly emerged variants as well as the infections of SARS-CoV-2 virus spike-protein of WT and its mutants-mediated pseudotyped viruses . EGCG shows maximum inhibitory effect against SARS-CoV-2 infection when the host cells are pre-incubated with the drug prior to viral infection. A sorbitol/lecithin-based throat spray containing concentrated green tea extract rich in EGCG content significantly reduces SARS-CoV-2 infectivity in oral mucosa. Several factors including degradation in gastrointestinal environment, low absorption in small intestine and extensive metabolism of EGCG are responsible for its poor bioavailability in humans. Pharmacokinetic and metabolism studies of EGCG in humans reveal poor bioavailability of EGCG in human plasma and EGCG-4"-sulfate is its major metabolite. The concentration of EGCG-4"-sulfate in human plasma is almost equivalent to that of free EGCG (Cmax 177.9 vs 233.5 nmol/L). These findings suggest that inhibition of sulfation of EGCG is a crucial factor for improvement of its bioavailability. In vitro study on the mechanism of EGCG sulfonation indicates that sulfotransferases, SULT1A1 and SULT1A3 are responsible for sulfonation in human liver and small intestine, respectively. Some attempts including structural modifications, and nanoformulations of EGCG and addition of nutrients with EGCG have been made to improve the bioavailability of EGCG. CONCLUSIONS The findings of this study suggest that EGCG has strong antiviral activity against SARS-CoV-2 infection independent of viral strains (Wuhan type (WT), other variants) by inhibition of viral entry and replication in host cells in vitro models. EGCG may be useful in reduction of this viral load in salivary glands of COVID-19 patients, if it is applied in mouth and throat wash formulations in optimal concentrations. EGCG could be a promising candidate in the development of effective vaccine for prevention of the infections of newly emergent strains of SARS-CoV-2 virus. EGCG might be useful also as a clinical medicine for treatment of COVID-19 patients if its bioavailability in human plasma is enhanced.
Collapse
Affiliation(s)
- Biswanath Dinda
- Department of Chemistry, Tripura University, Suryamaninagar, Agartala, Tripura, 799 022, India
| | - Subhajit Dinda
- Department of Chemistry, Kamalpur Govt Degree College, Dhalai,Tripura, 799 285, India
| | - Manikarna Dinda
- Department of Biochemistry and Molecular Genetics, University of Virginia, School of Medicine, Charlottesville, 1300 Jefferson Park Ave, VA, 22908, United States of America
| |
Collapse
|
9
|
Effects of Green Tea Polyphenol Epigallocatechin-3-Gallate on Markers of Inflammation and Fibrosis in a Rat Model of Pulmonary Silicosis. Int J Mol Sci 2023; 24:ijms24031857. [PMID: 36768179 PMCID: PMC9916388 DOI: 10.3390/ijms24031857] [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: 11/18/2022] [Revised: 01/08/2023] [Accepted: 01/14/2023] [Indexed: 01/19/2023] Open
Abstract
Inhalation of silica particles causes inflammatory changes leading to fibrotizing silicosis. Considering a lack of effective therapy, and a growing information on the wide actions of green tea polyphenols, particularly epigallocatechin-3-gallate (EGCG), the aim of this study was to evaluate the early effects of EGCG on markers of inflammation and lung fibrosis in silicotic rats. The silicosis model was induced by a single transoral intratracheal instillation of silica (50 mg/mL/animal), while controls received an equivalent volume of saline. The treatment with intraperitoneal EGCG (20 mg/kg, or saline in controls) was initiated the next day after silica instillation and was given twice a week. Animals were euthanized 14 or 28 days after the treatment onset, and the total and differential counts of leukocytes in the blood and bronchoalveolar lavage fluid (BALF), wet/dry lung weight ratio, and markers of inflammation, oxidative stress, and fibrosis in the lung were determined. The presence of collagen and smooth muscle mass in the walls of bronchioles and lung vessels was investigated immunohistochemically. Early treatment with EGCG showed some potential to alleviate inflammation, and a trend to decrease oxidative stress-induced changes, including apoptosis, and a prevention of fibrotic changes in the bronchioles and pulmonary vessels. However, further investigations should be undertaken to elucidate the effects of EGCG in the lung silicosis model in more detail. In addition, because of insufficient data from EGCG delivery in silicosis, the positive and eventual adverse effects of this herbal compound should be carefully studied before any preventive use or therapy with EGCG may be recommended.
Collapse
|
10
|
Therapeutic Effects of Green Tea Polyphenol (‒)-Epigallocatechin-3-Gallate (EGCG) in Relation to Molecular Pathways Controlling Inflammation, Oxidative Stress, and Apoptosis. Int J Mol Sci 2022; 24:ijms24010340. [PMID: 36613784 PMCID: PMC9820274 DOI: 10.3390/ijms24010340] [Citation(s) in RCA: 86] [Impact Index Per Article: 28.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2022] [Revised: 12/20/2022] [Accepted: 12/22/2022] [Indexed: 12/28/2022] Open
Abstract
(‒)-Epigallocatechin-3-gallate (EGCG) is the most abundant polyphenol in green tea. Thanks to multiple interactions with cell surface receptors, intracellular signaling pathways, and nuclear transcription factors, EGCG possesses a wide variety of anti-inflammatory, antioxidant, antifibrotic, anti-remodelation, and tissue-protective properties which may be useful in the treatment of various diseases, particularly in cancer, and neurological, cardiovascular, respiratory, and metabolic disorders. This article reviews current information on the biological effects of EGCG in the above-mentioned disorders in relation to molecular pathways controlling inflammation, oxidative stress, and cell apoptosis.
Collapse
|
11
|
Green Tea Polyphenol (-)-Epigallocatechin-3-Gallate (EGCG): A Time for a New Player in the Treatment of Respiratory Diseases? Antioxidants (Basel) 2022; 11:antiox11081566. [PMID: 36009285 PMCID: PMC9405266 DOI: 10.3390/antiox11081566] [Citation(s) in RCA: 48] [Impact Index Per Article: 16.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2022] [Revised: 08/03/2022] [Accepted: 08/11/2022] [Indexed: 12/13/2022] Open
Abstract
(-)-Epigallocatechin-3-gallate (EGCG) is a major polyphenol of green tea that possesses a wide variety of actions. EGCG acts as a strong antioxidant which effectively scavenges reactive oxygen species (ROS), inhibits pro-oxidant enzymes including NADPH oxidase, activates antioxidant systems including superoxide dismutase, catalase, or glutathione, and reduces abundant production of nitric oxide metabolites by inducible nitric oxide synthase. ECGC also exerts potent anti-inflammatory, anti-fibrotic, pro-apoptotic, anti-tumorous, and metabolic effects via modulation of a variety of intracellular signaling cascades. Based on this knowledge, the use of EGCG could be of benefit in respiratory diseases with acute or chronic inflammatory, oxidative, and fibrotizing processes in their pathogenesis. This article reviews current information on the biological effects of EGCG in those respiratory diseases or animal models in which EGCG has been administered, i.e., acute respiratory distress syndrome, respiratory infections, COVID-19, bronchial asthma, chronic obstructive pulmonary disease, lung fibrosis, silicosis, lung cancer, pulmonary hypertension, and lung embolism, and critically discusses effectiveness of EGCG administration in these respiratory disorders. For this review, articles in English language from the PubMed database were used.
Collapse
|
12
|
Rossi RE, Chen J, Caplin ME. The Role of Diet and Supplements in the Prevention and Progression of COVID-19: Current Knowledge and Open Issues. Prev Nutr Food Sci 2022; 27:137-149. [PMID: 35919576 PMCID: PMC9309075 DOI: 10.3746/pnf.2022.27.2.137] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2021] [Revised: 02/22/2022] [Accepted: 03/22/2022] [Indexed: 01/08/2023] Open
Abstract
A healthy diet and dietary supplements have gained attention as potential co-adjuvants in managing and preventing coronavirus disease 2019 (COVID-19). This paper critically reviews the current evidence regarding the impact of diet and supplements on the prevention and progression of COVID-19. According to available data, a healthy diet and normal weight are considered protective factors. Regarding dietary supplementation, the most robust results from human studies are for vitamin C, which appears to decrease inflammatory markers and suppress cytokine storm. A small, randomized trial showed that a high dose of vitamin D significantly reduced the need for intensive care unit treatment of patients requiring hospitalization for COVID-19. According to retrospective human studies, there is limited evidence for vitamin E and selenium supplements. Animal studies have investigated the effects of green tea and curcumin. Xanthohumol and probiotics, interesting for their antiviral, anti-inflammatory, and immunoregulatory properties, need formal clinical study. In summary, there is promising evidence supporting the role of diet and supplements as co-adjuvants in the treatment of COVID-19. Further studies and properly designed clinical trials are necessary to draw more robust conclusions; however, it is not unreasonable to take a pragmatic approach and promote the use of appropriate diet and supplements to counter the effects of COVID-19, ideally with a mechanism to assess outcomes.
Collapse
Affiliation(s)
- Roberta Elisa Rossi
- Hepatology and Hepato-Pancreatic-Biliary Surgery and Liver Transplantation, Fondazione IRCCS, Istituto Nazionale Tumori, Milan, MI 20133, Italy
- Department of Pathophysiology and Transplantation, University of Milan, Milan, MI 20122, Italy
| | - Jie Chen
- Department of Gastroenterology, First Affiliated Hospital, Sun Yat-Sen University, Guangzhou 510275, China
| | - Martyn Evan Caplin
- Centre for Gastroenterology, Royal Free Hospital, London NW3 2QG, UK
- Division of Medicine, Faculty of Medical Sciences, University College London, London WC1E 6BT, UK
| |
Collapse
|
13
|
Wang L, Zhu T, Feng D, Li R, Zhang C. Polyphenols from Chinese Herbal Medicine: Molecular Mechanisms and Therapeutic Targets in Pulmonary Fibrosis. THE AMERICAN JOURNAL OF CHINESE MEDICINE 2022; 50:1063-1094. [PMID: 35475972 DOI: 10.1142/s0192415x22500434] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
Pulmonary fibrosis (PF) is a highly confounding and fatal pathological process with finite treatment options. Multiple factors such as oxidative and immune/inflammation involve key pathological processes in chronic lung disease, and their intimate interactions mediate chronic lung damage, denudation of the alveolar epithelium, hyperproliferation of type II alveolar epithelial cells (AECIIs), proliferation and differentiation of fibroblasts, and the permeability of microvessels. We reviewed the classic mechanism of PF and highlighted a few emerging mechanisms for studying complex networks in lung disease pathology. Polyphenols, as a multi-target drug, has excellent potential in the treatment of pulmonary fibrosis. We then reviewed recent advances in discovering phenolic compounds from fruits, tea, and medical herbs with the bioactivities of simultaneously regulating multiple factors (e.g., oxidative stress, inflammation, autophagy, apoptosis, pyroptosis) for minimizing pulmonary fibrosis injury. These compounds include resveratrol, curcumin, salvianolic acid B, epigallocatechin-3-gallate, gallic acid, corilagin. Each phenolic compound can exert its anti-PF effect through various mechanisms, and the signaling pathways involved in different phenolic compounds are not the same. This review summarized the available evidence on phenolic compounds' effectiveness in pulmonary diseases and explored the molecular mechanisms and therapeutic targets of phenolic compounds from Chinese herbal medicine with the properties of inhibition of ongoing fibrogenesis and resolution of existing fibrosis.
Collapse
Affiliation(s)
- Lei Wang
- State Key Laboratory of Natural Medicines, School of Traditional Chinese Medicines, China Pharmaceutical University, Nanjing 210009, P. R. China.,Sino-Jan Joint Lab of Natural Health Products Research, School of Traditional Chinese Medicines, China Pharmaceutical University, Nanjing 210009, P. R. China
| | - Ting Zhu
- Institute of Neuroregeneration and Neurorehabilitation, Qingdao University, Qingdao 266071, P. R. China
| | - Deqin Feng
- State Key Laboratory of Microbial Resources, Chinese Academy of Sciences, Beijing 100049, P. R. China
| | - Renshi Li
- State Key Laboratory of Natural Medicines, School of Traditional Chinese Medicines, China Pharmaceutical University, Nanjing 210009, P. R. China.,Sino-Jan Joint Lab of Natural Health Products Research, School of Traditional Chinese Medicines, China Pharmaceutical University, Nanjing 210009, P. R. China
| | - Chaofeng Zhang
- State Key Laboratory of Natural Medicines, School of Traditional Chinese Medicines, China Pharmaceutical University, Nanjing 210009, P. R. China.,Sino-Jan Joint Lab of Natural Health Products Research, School of Traditional Chinese Medicines, China Pharmaceutical University, Nanjing 210009, P. R. China
| |
Collapse
|
14
|
Estornut C, Milara J, Bayarri MA, Belhadj N, Cortijo J. Targeting Oxidative Stress as a Therapeutic Approach for Idiopathic Pulmonary Fibrosis. Front Pharmacol 2022; 12:794997. [PMID: 35126133 PMCID: PMC8815729 DOI: 10.3389/fphar.2021.794997] [Citation(s) in RCA: 59] [Impact Index Per Article: 19.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2021] [Accepted: 12/10/2021] [Indexed: 01/19/2023] Open
Abstract
Idiopathic pulmonary fibrosis (IPF) is a chronic interstitial lung disease characterized by an abnormal reepithelialisation, an excessive tissue remodelling and a progressive fibrosis within the alveolar wall that are not due to infection or cancer. Oxidative stress has been proposed as a key molecular process in pulmonary fibrosis development and different components of the redox system are altered in the cellular actors participating in lung fibrosis. To this respect, several activators of the antioxidant machinery and inhibitors of the oxidant species and pathways have been assayed in preclinical in vitro and in vivo models and in different clinical trials. This review discusses the role of oxidative stress in the development and progression of IPF and its underlying mechanisms as well as the evidence of oxidative stress in human IPF. Finally, we analyze the mechanism of action, the efficacy and the current status of different drugs developed to inhibit the oxidative stress as anti-fibrotic therapy in IPF.
Collapse
Affiliation(s)
- Cristina Estornut
- Department of Pharmacology, Faculty of Medicine, University of Valencia, Valencia, Spain
- *Correspondence: Cristina Estornut, ; Javier Milara,
| | - Javier Milara
- Department of Pharmacology, Faculty of Medicine, University of Valencia, Valencia, Spain
- Pharmacy Unit, University General Hospital Consortium, Valencia, Spain
- CIBERES, Health Institute Carlos III, Valencia, Spain
- *Correspondence: Cristina Estornut, ; Javier Milara,
| | - María Amparo Bayarri
- Department of Pharmacology, Faculty of Medicine, University of Valencia, Valencia, Spain
| | - Nada Belhadj
- Department of Pharmacology, Faculty of Medicine, University of Valencia, Valencia, Spain
| | - Julio Cortijo
- Department of Pharmacology, Faculty of Medicine, University of Valencia, Valencia, Spain
- Pharmacy Unit, University General Hospital Consortium, Valencia, Spain
- CIBERES, Health Institute Carlos III, Valencia, Spain
- Research and Teaching Unit, University General Hospital Consortium, Valencia, Spain
| |
Collapse
|
15
|
Yao JJ, Ma QQ, Shen WW, Li LC, Hu D. Nano-enabled delivery of EGCG ameliorates silica-induced pulmonary fibrosis in rats. Toxicology 2022; 469:153114. [DOI: 10.1016/j.tox.2022.153114] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2021] [Revised: 01/21/2022] [Accepted: 01/30/2022] [Indexed: 01/23/2023]
|
16
|
Wang Y, He Y, Rayman MP, Zhang J. Prospective Selective Mechanism of Emerging Senolytic Agents Derived from Flavonoids. JOURNAL OF AGRICULTURAL AND FOOD CHEMISTRY 2021; 69:12418-12423. [PMID: 34662116 DOI: 10.1021/acs.jafc.1c04379] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/13/2023]
Abstract
Senescent cells (SCs) are associated with the onset and development of multiple chronic diseases. Selective clearance of SCs by senolytic drugs is a potential therapeutic option for a number of age-related diseases. Among senolytic candidates, only dasatinib with quercetin and fisetin meet the rigorous criteria for senolytic drugs, according to a modified version of Koch's postulates. It is astonishing that two of the three agents, i.e., quercetin and fisetin, are flavonoids, although the mechanism by which they preferentially eliminate SCs is unclear. Herein, we propose a possible selective mechanism; prooxidant activities of quercetin or fisetin are inevitably involved in killing apoptosis-resistant SCs. Among the dietary flavonoids, quercetin is a potent redox-active flavonoid with strong prooxidant activities, and transition metals, such as copper and iron, hugely amplify its prooxidant activities. Fisetin, which has higher senolytic activities than quercetin, has higher prooxidant effects than quercetin in the absence or presence of copper. It appears that the prooxidant activity of flavonoids is an important consideration for screening senolytics. SCs accumulate high levels of copper and iron, and the selective mechanism of quercetin or fisetin is probably associated with copper/iron-promoted oxidative damage in SCs. Copper and iron dramatically enhanced the prooxidant effects of the flavonoid, epigallocatechin-3-gallate, having shown a depletion effect on SCs in rats and high therapeutic efficacy in patients with idiopathic pulmonary fibrosis, largely caused by SCs. Further investigation to evaluate whether epigallocatechin-3-gallate is a senolytic drug, according to Koch's postulates, is warranted.
Collapse
Affiliation(s)
- Yijun Wang
- State Key Laboratory of Tea Plant Biology and Utilization, School of Tea & Food Science, Anhui Agricultural University, Hefei, Anhui 230036, People's Republic of China
| | - Yufeng He
- State Key Laboratory of Tea Plant Biology and Utilization, School of Tea & Food Science, Anhui Agricultural University, Hefei, Anhui 230036, People's Republic of China
| | - Margaret P Rayman
- Department of Nutritional Sciences, Faculty of Health and Medical Sciences, University of Surrey, Guildford GU2 7XH, United Kingdom
| | - Jinsong Zhang
- State Key Laboratory of Tea Plant Biology and Utilization, School of Tea & Food Science, Anhui Agricultural University, Hefei, Anhui 230036, People's Republic of China
| |
Collapse
|
17
|
Zhang Z, Zhang X, Bi K, He Y, Yan W, Yang CS, Zhang J. Potential protective mechanisms of green tea polyphenol EGCG against COVID-19. Trends Food Sci Technol 2021; 114:11-24. [PMID: 34054222 PMCID: PMC8146271 DOI: 10.1016/j.tifs.2021.05.023] [Citation(s) in RCA: 82] [Impact Index Per Article: 20.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2021] [Revised: 05/07/2021] [Accepted: 05/15/2021] [Indexed: 02/08/2023]
Abstract
Background The world is in the midst of the COVID-19 pandemic. In this comprehensive review, we discuss the potential protective effects of (−)-epigallocatechin-3-gallate (EGCG), a major constituent of green tea, against COVID-19. Scope and approach Information from literature of clinical symptoms and molecular pathology of COVID-19 as well as relevant publications in which EGCG shows potential protective activities against COVID-19 is integrated and evaluated. Key findings and conclusions EGCG, via activating Nrf2, can suppress ACE2 (a cellular receptor for SARS-CoV-2) and TMPRSS2, which mediate cell entry of the virus. Through inhibition of SARS-CoV-2 main protease, EGCG may inhibit viral reproduction. EGCG via its broad antioxidant activity may protect against SARS-CoV-2 evoked mitochondrial ROS (which promote SARS-CoV-2 replication) and against ROS burst inflicted by neutrophil extracellular traps. By suppressing ER-resident GRP78 activity and expression, EGCG can potentially inhibit SARS-CoV-2 life cycle. EGCG also shows protective effects against 1) cytokine storm-associated acute lung injury/acute respiratory distress syndrome, 2) thrombosis via suppressing tissue factors and activating platelets, 3) sepsis by inactivating redox-sensitive HMGB1, and 4) lung fibrosis through augmenting Nrf2 and suppressing NF-κB. These activities remain to be further substantiated in animals and humans. The possible concerted actions of EGCG suggest the importance of further studies on the prevention and treatment of COVID-19 in humans. These results also call for epidemiological studies on potential preventive effects of green tea drinking on COVID-19.
Collapse
Affiliation(s)
- Zhichao Zhang
- Department of Musculoskeletal Tumor, Fudan University Shanghai Cancer Center, Shanghai, 200032, China
| | - Xiangchun Zhang
- Tea Research Institute, Chinese Academy of Agricultural Sciences, Hangzhou, 310008, China
| | - Keyi Bi
- State Key Laboratory of Tea Plant Biology and Utilization, School of Tea & Food Science, Anhui Agricultural University, Hefei, 230036, China
| | - Yufeng He
- State Key Laboratory of Tea Plant Biology and Utilization, School of Tea & Food Science, Anhui Agricultural University, Hefei, 230036, China
| | - Wangjun Yan
- Department of Musculoskeletal Tumor, Fudan University Shanghai Cancer Center, Shanghai, 200032, China
| | - Chung S Yang
- Department of Chemical Biology, Ernest Mario School of Pharmacy, Rutgers, The State University of New Jersey, Piscataway, NJ, 08854-8020, USA
| | - Jinsong Zhang
- State Key Laboratory of Tea Plant Biology and Utilization, School of Tea & Food Science, Anhui Agricultural University, Hefei, 230036, China
| |
Collapse
|
18
|
Rajasekaran S, Rajasekar N, Sivanantham A. Therapeutic potential of plant-derived tannins in non-malignant respiratory diseases. J Nutr Biochem 2021; 94:108632. [PMID: 33794331 DOI: 10.1016/j.jnutbio.2021.108632] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2020] [Revised: 01/06/2021] [Accepted: 03/23/2021] [Indexed: 12/24/2022]
Abstract
Respiratory diseases are the major cause of human illness and death around the world. Despite advances in detection and treatment, very few classes of safe and effective therapy have been introduced to date. At present, phytochemicals are getting more attention because of their diverse beneficial activities and minimal toxicity. Tannins are polyphenolic secondary metabolites with high molecular weights, which are naturally present in a wide variety of fruits, vegetables, cereals, and leguminous seeds. Many tannins are endowed with well-recognized protective properties, such as anti-cancer, anti-microbial, anti-oxidant, anti-hyperglycemic, and many others. This review summarizes a large body of experimental evidence implicating that tannins are helpful in tackling a wide range of non-malignant respiratory diseases including acute lung injury (ALI), pulmonary fibrosis, asthma, pulmonary hypertension, and chronic obstructive pulmonary disease (COPD). Mechanistic pathways by which various classes of tannins execute their beneficial effects are discussed. In addition, clinical trials and our perspective on future research with tannins are also reviewed.
Collapse
Affiliation(s)
- Subbiah Rajasekaran
- Department of Biochemistry, ICMR-National Institute for Research in Environmental Health, Bhopal, Madhya Pradesh, India.
| | - Nandhine Rajasekar
- Department of Biotechnology, BIT-Campus, Anna University, Tiruchirappalli, Tamil Nadu, India
| | - Ayyanar Sivanantham
- Department of Biotechnology, BIT-Campus, Anna University, Tiruchirappalli, Tamil Nadu, India
| |
Collapse
|
19
|
Dutka M, Bobiński R, Ulman-Włodarz I, Hajduga M, Bujok J, Pająk C, Ćwiertnia M. Various aspects of inflammation in heart failure. Heart Fail Rev 2021; 25:537-548. [PMID: 31705352 PMCID: PMC7181445 DOI: 10.1007/s10741-019-09875-1] [Citation(s) in RCA: 44] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/08/2023]
Abstract
Despite significant advances in the prevention and treatment of heart failure (HF), the prognosis in patients who have been hospitalised on at least one occasion due to exacerbation of HF is still poor. Therefore, a better understanding of the underlying pathophysiological mechanisms of HF is crucial in order to achieve better results in the treatment of this clinical syndrome. One of the areas that, for years, has aroused the interest of researchers is the activation of the immune system and the elevated levels of biomarkers of inflammation in patients with both ischaemic and non-ischaemic HF. Additionally, it is intriguing that the level of circulating pro-inflammatory biomarkers correlates with the severity of the disease and prognosis in this group of patients. Unfortunately, clinical trials aimed at assessing interventions to modulate the inflammatory response in HF have been disappointing, and the modulation of the inflammatory response has had either no effect or even a negative effect on the HF prognosis. The article presents a summary of current knowledge on the role of immune system activation and inflammation in the pathogenesis of HF. Understanding the immunological mechanisms pathogenetically associated with left ventricular remodelling and progression of HF may open up new therapeutic possibilities for HF.
Collapse
Affiliation(s)
- Mieczysław Dutka
- Faculty of Health Sciences, Department of Biochemistry and Molecular Biology, University of Bielsko-Biala, Willowa St. 2, 43-309, Bielsko-Biala, Poland.
| | - Rafał Bobiński
- Faculty of Health Sciences, Department of Biochemistry and Molecular Biology, University of Bielsko-Biala, Willowa St. 2, 43-309, Bielsko-Biala, Poland
| | - Izabela Ulman-Włodarz
- Faculty of Health Sciences, Department of Biochemistry and Molecular Biology, University of Bielsko-Biala, Willowa St. 2, 43-309, Bielsko-Biala, Poland
| | - Maciej Hajduga
- Faculty of Health Sciences, Department of Biochemistry and Molecular Biology, University of Bielsko-Biala, Willowa St. 2, 43-309, Bielsko-Biala, Poland
| | - Jan Bujok
- Faculty of Health Sciences, Department of Biochemistry and Molecular Biology, University of Bielsko-Biala, Willowa St. 2, 43-309, Bielsko-Biala, Poland
| | - Celina Pająk
- Faculty of Health Sciences, Department of Biochemistry and Molecular Biology, University of Bielsko-Biala, Willowa St. 2, 43-309, Bielsko-Biala, Poland
| | - Michał Ćwiertnia
- Faculty of Health Sciences, Department of Emergency Medicine, University of Bielsko-Biala, Willowa St. 2, 43-309, Bielsko-Biala, Poland
| |
Collapse
|
20
|
Shiha G, Soliman R, Elbasiony M, Darwish NHE, Mousa SA. Novel combined single dose anti-hepatitis C therapy: a pilot study. Sci Rep 2021; 11:4623. [PMID: 33633233 PMCID: PMC7907074 DOI: 10.1038/s41598-021-84066-3] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2020] [Accepted: 02/09/2021] [Indexed: 12/15/2022] Open
Abstract
The new anti-hepatitis C virus (HCV) molecules improve treatment regimens and outcomes, but there are drawbacks. New combinations should target the HCV infectious cycle and be effective against all HCV genotypes. We developed the novel formulation Catvira, composed of epigallocatechingallate (EGCG) + sofosbuvir + ribavirin. Here, we compared Catvira to sofosbuvir + ribavirin tablets in patients with CHC genotype 4 in a randomized open-label efficacy and safety study. Treatment-naïve and treatment-experienced patients (n = 80) were randomly assigned to receive a single daily fixed dose of Catvira or sofosbuvir + ribavirin for 12 or 24 weeks. Both Catvira and sofosbuvir + ribavirin yielded similar outcomes of viral load (p < 0.001). Patients receiving Catvira had a significantly more rapid rate of viral load decline with sustained virologic response (SVR12) achieved by 90% of patients receiving 12 weeks of treatment. Catvira did not impact hemoglobin levels while sofosbuvir + ribavirin showed significant decline in hemoglobin levels after 24 weeks (p < 0.05). In this clinical trial (ClinicalTrials.gov Identifier NCT02483156), we found that Catvira administered daily for 12 or 24 weeks is safe, effective, and well-tolerated in both naïve and treatment-experienced patients with HCV genotype 4.
Collapse
Affiliation(s)
- Gamal Shiha
- Internal Medicine Department, Faculty of Medicine, Mansoura University, Mansoura, Egypt
- Egyptian Liver Research Institute and Hospital (ELRIAH), Mansoura, Egypt
| | - Reham Soliman
- Egyptian Liver Research Institute and Hospital (ELRIAH), Mansoura, Egypt
- Tropical Medicine Department, Faculty of Medicine, Port Said University, Port Said, Egypt
| | - Mohamed Elbasiony
- Internal Medicine Department, Faculty of Medicine, Mansoura University, Mansoura, Egypt
- Egyptian Liver Research Institute and Hospital (ELRIAH), Mansoura, Egypt
| | - Noureldien H E Darwish
- Hematology Unit, Clinical Pathology Department, Faculty of Medicine, Mansoura University, Mansoura, Egypt
- The Pharmaceutical Research Institute, Albany College of Pharmacy and Health Sciences, 1 Discovery Drive, Rensselaer, NY, 12144, USA
| | - Shaker A Mousa
- The Pharmaceutical Research Institute, Albany College of Pharmacy and Health Sciences, 1 Discovery Drive, Rensselaer, NY, 12144, USA.
- Virothera Pharmaceuticals, Rensselaer, NY, USA.
| |
Collapse
|
21
|
Dinesh Babu V, Suresh Kumar A, Sudhandiran G. Diosgenin inhibits TGF-β1/Smad signaling and regulates epithelial mesenchymal transition in experimental pulmonary fibrosis. Drug Chem Toxicol 2020; 45:1264-1275. [PMID: 32924642 DOI: 10.1080/01480545.2020.1814803] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/24/2023]
Abstract
Idiopathic Pulmonary Fibrosis (IPF) is a grave disease characterized by abnormal wound healing associated with chronic, progressive, irreversible fatal lung disease, leading to persistent injuries to the alveolar epithelium. A consequent disturbance of fibroblast proliferation and apoptosis results in subsequent release of pro-inflammatory and pro-fibrotic mediators coupled with accumulation of extracellular matrix within the interstitium. Inexorable distortion of lung alveolar architecture leads to respiratory failure with a median survival rate of 3-5 years. Currently available drugs can only slowdown the progression of fibrosis and novel drugs are warranted to treat this disease. In this study, we demonstrate the fibro-protective effect of diosgenin in experimental lung fibrosis through regulation of Epithelial Mesenchymal Transition (EMT). A single dose of 3 U/kg body weight (b.wt) Bleomycin (BLM) was administered intratracheally in Wistar male albino rats and fibrotic animals were treated with diosgenin (100 mg/kg b.wt) orally for 28 days. BLM administered rat show histological alteration with increased mast cell and collagen accumulation. BLM induced abnormalities were significantly reduced upon treatment with diosgenin. Western blot analysis revealed an increased level of pro-inflammatory and pro-fibrotic molecules such as IL-1β and TGF-β in BLM induced rats. Rats supplemented with diosgenin showed a decreased expression of inflammatory and pro-fibrotic mediators. Markers of EMT molecules were evaluated by immunoblot. The results of immunoblot demonstrate that diosgenin regulated the expression of TGF-β mediated EMT. Hence, from the overall study, administration of diosgenin prevents pulmonary fibrosis by restraint inflammation and EMT.
Collapse
Affiliation(s)
- Vadivel Dinesh Babu
- Department of Biochemistry, Cell Biology Laboratory, University of Madras, Chennai, India
| | | | - Ganapasam Sudhandiran
- Department of Biochemistry, Cell Biology Laboratory, University of Madras, Chennai, India
| |
Collapse
|
22
|
Insights into the Role of Bioactive Food Ingredients and the Microbiome in Idiopathic Pulmonary Fibrosis. Int J Mol Sci 2020; 21:ijms21176051. [PMID: 32842664 PMCID: PMC7503951 DOI: 10.3390/ijms21176051] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2020] [Revised: 08/18/2020] [Accepted: 08/19/2020] [Indexed: 02/08/2023] Open
Abstract
Idiopathic pulmonary fibrosis (IPF) is a chronic disease mainly associated with aging and, to date, its causes are still largely unknown. It has been shown that dietary habits can accelerate or delay the occurrence of aging-related diseases; however, their potential role in IPF development has been underestimated so far. The present review summarizes the evidence regarding the relationship between diet and IPF in humans, and in animal models of pulmonary fibrosis, in which we discuss the bioactivity of specific dietary food ingredients, including fatty acids, peptides, amino acids, carbohydrates, vitamins, minerals and phytochemicals. Interestingly, many animal studies reveal preventive and therapeutic effects of particular compounds. Furthermore, it has been recently suggested that the lung and gut microbiota could be involved in IPF, a relationship which may be linked to changes in immunological and inflammatory factors. Thus, all the evidence so far puts forward the idea that the gut-lung axis could be modulated by dietary factors, which in turn have an influence on IPF development. Overall, the data reviewed here support the notion of identifying food ingredients with potential benefits in IPF, with the ultimate aim of designing nutritional approaches as an adjuvant therapeutic strategy.
Collapse
|
23
|
Menegazzi M, Campagnari R, Bertoldi M, Crupi R, Di Paola R, Cuzzocrea S. Protective Effect of Epigallocatechin-3-Gallate (EGCG) in Diseases with Uncontrolled Immune Activation: Could Such a Scenario Be Helpful to Counteract COVID-19? Int J Mol Sci 2020; 21:ijms21145171. [PMID: 32708322 PMCID: PMC7404268 DOI: 10.3390/ijms21145171] [Citation(s) in RCA: 68] [Impact Index Per Article: 13.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2020] [Revised: 07/16/2020] [Accepted: 07/18/2020] [Indexed: 01/22/2023] Open
Abstract
Some coronavirus disease 2019 (COVID-19) patients develop acute pneumonia which can result in a cytokine storm syndrome in response to Severe Acute Respiratory Syndrome coronavirus 2 (SARS-CoV-2) infection. The most effective anti-inflammatory drugs employed so far in severe COVID-19 belong to the cytokine-directed biological agents, widely used in the management of many autoimmune diseases. In this paper we analyze the efficacy of epigallocatechin 3-gallate (EGCG), the most abundant ingredient in green tea leaves and a well-known antioxidant, in counteracting autoimmune diseases, which are dominated by a massive cytokines production. Indeed, many studies registered that EGCG inhibits signal transducer and activator of transcription (STAT)1/3 and nuclear factor kappa-light-chain-enhancer of activated B cells (NF-κB) transcription factors, whose activities are crucial in a multiplicity of downstream pro-inflammatory signaling pathways. Importantly, the safety of EGCG/green tea extract supplementation is well documented in many clinical trials, as discussed in this review. Since EGCG can restore the natural immunological homeostasis in many different autoimmune diseases, we propose here a supplementation therapy with EGCG in COVID-19 patients. Besides some antiviral and anti-sepsis actions, the major EGCG benefits lie in its anti-fibrotic effect and in the ability to simultaneously downregulate expression and signaling of many inflammatory mediators. In conclusion, EGCG can be considered a potential safe natural supplement to counteract hyper-inflammation growing in COVID-19.
Collapse
Affiliation(s)
- Marta Menegazzi
- Department of Neuroscience, Biomedicine and Movement Sciences, Biochemistry Section, School of Medicine, University of Verona, Strada Le Grazie 8, I-37134 Verona, Italy; (R.C.); (M.B.)
- Correspondence:
| | - Rachele Campagnari
- Department of Neuroscience, Biomedicine and Movement Sciences, Biochemistry Section, School of Medicine, University of Verona, Strada Le Grazie 8, I-37134 Verona, Italy; (R.C.); (M.B.)
| | - Mariarita Bertoldi
- Department of Neuroscience, Biomedicine and Movement Sciences, Biochemistry Section, School of Medicine, University of Verona, Strada Le Grazie 8, I-37134 Verona, Italy; (R.C.); (M.B.)
| | - Rosalia Crupi
- Department of Veterinary Science, University of Messina, Polo Universitario dell’Annunziata, I-98168 Messina, Italy;
| | - Rosanna Di Paola
- Department of Chemical, Biological, Pharmaceutical and Environmental Sciences, University of Messina, Viale Ferdinando Stagno D’Alcontres 31, I-98166 Messina, Italy; (R.D.P.); (S.C.)
| | - Salvatore Cuzzocrea
- Department of Chemical, Biological, Pharmaceutical and Environmental Sciences, University of Messina, Viale Ferdinando Stagno D’Alcontres 31, I-98166 Messina, Italy; (R.D.P.); (S.C.)
- Department of Pharmacological and Physiological Science, Saint Louis University School of Medicine, St. Louis, MO 63104, USA
| |
Collapse
|
24
|
Zhang Q, Gan C, Liu H, Wang L, Li Y, Tan Z, You J, Yao Y, Xie Y, Yin W, Ye T. Cryptotanshinone reverses the epithelial-mesenchymal transformation process and attenuates bleomycin-induced pulmonary fibrosis. Phytother Res 2020; 34:2685-2696. [PMID: 32281701 DOI: 10.1002/ptr.6699] [Citation(s) in RCA: 20] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2019] [Revised: 03/13/2020] [Accepted: 03/25/2020] [Indexed: 02/05/2023]
Abstract
Idiopathic pulmonary fibrosis (IPF) is a fibrotic interstitial pneumonia that causes pulmonary tissue damage and functional impairment. To investigate the effects of cryptotanshinone on pulmonary fibrosis, the expression of NIH/3T3, HPF, and rat primary pulmonary fibroblasts was measured and found to be inhibited by CPT in a time- and concentration-dependent manner, and the upregulation of α-SMA expression in NIH/3T3 and HPF cells, which had been stimulated by TGFβ-1, was decreased after CPT administration. We observed that CPT could reverse the increase in α-SMA expression and vimentin and the decrease in E-cad expression in A549 cells, which had been induced by 5 ng/mL TGFβ-1, indicating that CPT has inhibitory effects in the EMT process. A BLM-induced pulmonary fibrosis model was established in C57BL/6 mice. The lung coefficient and hydroxyproline content increased significantly in the BLM-induced group and were decreased in the CPT-treated group. The expression levels of collagen-I and α-SMA and the phosphorylation level of Stat3 were significantly increased, and CPT treatment decreased these levels. Furthermore, the results from the flow cytometry analysis indicated that, in lung tissues, the frequencies of MDSCs, macrophages, DCs and T cells were considerably increased in the BLM-induced group, while CPT treatment reduced these immunocyte populations.
Collapse
Affiliation(s)
- Qianyu Zhang
- Department of Liver Surgery & Liver Transplantation, State Key of Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University and Collaborative Innovation Center for Biotherapy, Chengdu, Sichuan, People's Republic of China.,West China School of Public Health and Heathy Food Evaluation Research Center and West China Fourth Hospital, Sichuan University, Chengdu, Sichuan, People's Republic of China
| | - Cailing Gan
- Department of Liver Surgery & Liver Transplantation, State Key of Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University and Collaborative Innovation Center for Biotherapy, Chengdu, Sichuan, People's Republic of China
| | - Hongyao Liu
- Department of Liver Surgery & Liver Transplantation, State Key of Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University and Collaborative Innovation Center for Biotherapy, Chengdu, Sichuan, People's Republic of China
| | - Liqun Wang
- West China School of Public Health and Heathy Food Evaluation Research Center and West China Fourth Hospital, Sichuan University, Chengdu, Sichuan, People's Republic of China
| | - Yali Li
- West China School of Public Health and Heathy Food Evaluation Research Center and West China Fourth Hospital, Sichuan University, Chengdu, Sichuan, People's Republic of China
| | - Zui Tan
- Department of Liver Surgery & Liver Transplantation, State Key of Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University and Collaborative Innovation Center for Biotherapy, Chengdu, Sichuan, People's Republic of China
| | - Jia You
- West China School of Public Health and Heathy Food Evaluation Research Center and West China Fourth Hospital, Sichuan University, Chengdu, Sichuan, People's Republic of China
| | - Yuqin Yao
- West China School of Public Health and Heathy Food Evaluation Research Center and West China Fourth Hospital, Sichuan University, Chengdu, Sichuan, People's Republic of China
| | - Yongmei Xie
- Department of Liver Surgery & Liver Transplantation, State Key of Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University and Collaborative Innovation Center for Biotherapy, Chengdu, Sichuan, People's Republic of China
| | - Wenya Yin
- West China School of Public Health and Heathy Food Evaluation Research Center and West China Fourth Hospital, Sichuan University, Chengdu, Sichuan, People's Republic of China
| | - Tinghong Ye
- Department of Liver Surgery & Liver Transplantation, State Key of Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University and Collaborative Innovation Center for Biotherapy, Chengdu, Sichuan, People's Republic of China
| |
Collapse
|
25
|
Mahalanobish S, Saha S, Dutta S, Sil PC. Matrix metalloproteinase: An upcoming therapeutic approach for idiopathic pulmonary fibrosis. Pharmacol Res 2020; 152:104591. [PMID: 31837390 DOI: 10.1016/j.phrs.2019.104591] [Citation(s) in RCA: 75] [Impact Index Per Article: 15.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/20/2019] [Revised: 12/04/2019] [Accepted: 12/06/2019] [Indexed: 01/26/2023]
Abstract
Idiopathic pulmonary fibrosis (IPF) is a debilitating condition where excess collagen deposition occurs in the extracellular matrix. At first sight, it is expected that the level of different kinds of matrix metalloproteinases might be downregulated in IPF as it is a matrix degrading collagenase. However, the role of some matrix metalloproteinases (MMPs) is profibrotic where others have anti-fibrotic functions. These profibrotic MMPs effectively promote fibrosis development by stimulating the process of epithelial to mesenchymal transition. These profibrotic groups also induce macrophage polarization and fibrocyte migration. All of these events ultimately disrupt the balance between profibrotic and antifibrotic mediators, resulting aberrant repair process. Therefore, inhibition of these matrix metalloproteinases functions in IPF is a potential therapeutic approach. In addition to the use of synthetic inhibitor, various natural compounds, gene silencing act as potential natural MMP inhibitor to recover IPF.
Collapse
Affiliation(s)
- Sushweta Mahalanobish
- Division of Molecular Medicine, Bose Institute, P-1/12, CIT Scheme VII M, Kolkata, 700054, India
| | - Sukanya Saha
- Division of Molecular Medicine, Bose Institute, P-1/12, CIT Scheme VII M, Kolkata, 700054, India
| | - Sayanta Dutta
- Division of Molecular Medicine, Bose Institute, P-1/12, CIT Scheme VII M, Kolkata, 700054, India
| | - Parames C Sil
- Division of Molecular Medicine, Bose Institute, P-1/12, CIT Scheme VII M, Kolkata, 700054, India.
| |
Collapse
|
26
|
Xiang L, Zhang Q, Chi C, Wu G, Lin Z, Li J, Gu Q, Chen G. Curcumin analog A13 alleviates oxidative stress by activating Nrf2/ARE pathway and ameliorates fibrosis in the myocardium of high-fat-diet and streptozotocin-induced diabetic rats. Diabetol Metab Syndr 2020; 12:1. [PMID: 31921358 PMCID: PMC6947902 DOI: 10.1186/s13098-019-0485-z] [Citation(s) in RCA: 40] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/18/2019] [Accepted: 10/17/2019] [Indexed: 11/25/2022] Open
Abstract
BACKGROUND Diabetes mellitus is an important risk factor for cardiomyopathy. Increasing oxidative stress may be one of the main factors of diabetic cardiomyopathy. A13, a newly synthesized curcumin analog, was proved to be superior to curcumin in biological activity. However, little know about how A13 performed in diabetic models. In this study, we evaluated the ability of curcumin analog A13 to alleviate oxidative stress and ameliorate fibrosis in the myocardium, and explore the underlying mechanisms. METHODS Intraperitoneal injection of streptozotocin (30 mg/kg in 0.1 M sodium citrate buffer, pH 4.5) induced diabetes in high-fat fed rats. The rats were respectively treated with a daily dose of curcumin or A13 via intragastric intubation for 8 weeks. Myocardial tissue sections were stained with hematoxylin-eosin; oxidative stress was detected by biochemical assays; activation of the Nrf2/ARE pathway was detected by Western blot, immunohistochemical staining and RT-qPCR; myocardial fibrosis was identified by Western blot and Masson trichrome staining. RESULTS Treatment with curcumin analog A13 reduced the histological lesions of the myocardium in diabetic rats. Curcumin and A13 treatment decreased the malondialdehyde level and increased the activity of superoxide dismutase in the myocardium of diabetic rats. Molecular analysis and immunohistochemical staining demonstrated that dose of 20 mg/kg of A13 could activate the Nrf2/ARE pathway. Molecular analysis and Masson staining showed that curcumin analog A13 treatment significantly ameliorated fibrosis in myocardium of these diabetic rats. CONCLUSION Treatment with curcumin analog A13 protects the morphology of myocardium, restores the MDA levels and SOD activity, activates the Nrf2/ARE pathway and ameliorates myocardial fibrosis in diabetic rats. It may be a useful therapeutic agent for some aspects of diabetic cardiomyopathy.
Collapse
Affiliation(s)
- Lanting Xiang
- Department of Pathology, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang People’s Republic of China
| | - Qiongying Zhang
- Department of Pathology, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang People’s Republic of China
| | - Chen Chi
- Department of Pathology, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang People’s Republic of China
| | - Gu Wu
- Department of Pathology, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang People’s Republic of China
| | - Zhongmin Lin
- Department of Pathology, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang People’s Republic of China
| | - Jianmin Li
- Department of Pathology, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang People’s Republic of China
| | - Qianru Gu
- Department of Pathology, Sir Run Run Shaw Hospital affiliated To Zhejiang University School of Medicine, Hangzhou, Zhejiang People’s Republic of China
| | - Guorong Chen
- Department of Pathology, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang People’s Republic of China
| |
Collapse
|
27
|
Tsai MJ, Chang WA, Liao SH, Chang KF, Sheu CC, Kuo PL. The Effects of Epigallocatechin Gallate (EGCG) on Pulmonary Fibroblasts of Idiopathic Pulmonary Fibrosis (IPF)-A Next-Generation Sequencing and Bioinformatic Approach. Int J Mol Sci 2019; 20:E1958. [PMID: 31013581 PMCID: PMC6514693 DOI: 10.3390/ijms20081958] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2019] [Revised: 04/12/2019] [Accepted: 04/18/2019] [Indexed: 12/19/2022] Open
Abstract
Idiopathic pulmonary fibrosis (IPF) is a disabling and lethal chronic progressive pulmonary disease. Epigallocatechin gallate (EGCG) is a polyphenol, which is the major biological component of green tea. The anti-oxidative, anti-inflammatory, and anti-fibrotic effects of EGCG have been shown in some studies, whereas its effects in altering gene expression in pulmonary fibroblasts have not been systematically investigated. This study aimed to explore the effect of EGCG on gene expression profiles in fibroblasts of IPF. The pulmonary fibroblasts from an IPF patient were treated with either EGCG or water, and the expression profiles of mRNAs and microRNAs were determined by next-generation sequencing (NGS) and analyzed with the bioinformatics approach. A total of 61 differentially expressed genes and 56 differentially expressed microRNAs were found in EGCG-treated IPF fibroblasts. Gene ontology analyses revealed that the differentially expressed genes were mainly involved in the biosynthetic and metabolic processes of cholesterol. In addition, five potential altered microRNA-mRNA interactions were found, including hsa-miR-939-5p-PLXNA4, hsa-miR-3918-CTIF, hsa-miR-4768-5p-PDE5A, hsa-miR-1273g-3p-VPS53, and hsa-miR-1972-PCSK9. In summary, differentially expressed genes and microRNAs in response to EGCG treatment in IPF fibroblasts were identified in the current study. Our findings provide a scientific basis to evaluate the potential benefits of EGCG in IPF treatment, and warrant future studies to understand the role of molecular pathways underlying cholesterol homeostasis in the pathogenesis of IPF.
Collapse
Affiliation(s)
- Ming-Ju Tsai
- Graduate Institute of Clinical Medicine, College of Medicine, Kaohsiung Medical University, Kaohsiung 807, Taiwan.
- Division of Pulmonary and Critical Care Medicine, Department of Internal Medicine, Kaohsiung Medical University Hospital, Kaohsiung Medical University, Kaohsiung 807, Taiwan.
- Department of Internal Medicine, School of Medicine, College of Medicine, Kaohsiung Medical University, Kaohsiung 807, Taiwan.
- Department of Respiratory Therapy, School of Medicine, College of Medicine, Kaohsiung Medical University, Kaohsiung 807, Taiwan.
| | - Wei-An Chang
- Graduate Institute of Clinical Medicine, College of Medicine, Kaohsiung Medical University, Kaohsiung 807, Taiwan.
- Division of Pulmonary and Critical Care Medicine, Department of Internal Medicine, Kaohsiung Medical University Hospital, Kaohsiung Medical University, Kaohsiung 807, Taiwan.
- Department of Internal Medicine, School of Medicine, College of Medicine, Kaohsiung Medical University, Kaohsiung 807, Taiwan.
| | - Ssu-Hui Liao
- Graduate Institute of Clinical Medicine, College of Medicine, Kaohsiung Medical University, Kaohsiung 807, Taiwan.
| | | | - Chau-Chyun Sheu
- Graduate Institute of Clinical Medicine, College of Medicine, Kaohsiung Medical University, Kaohsiung 807, Taiwan.
- Division of Pulmonary and Critical Care Medicine, Department of Internal Medicine, Kaohsiung Medical University Hospital, Kaohsiung Medical University, Kaohsiung 807, Taiwan.
- Department of Internal Medicine, School of Medicine, College of Medicine, Kaohsiung Medical University, Kaohsiung 807, Taiwan.
- Department of Respiratory Therapy, School of Medicine, College of Medicine, Kaohsiung Medical University, Kaohsiung 807, Taiwan.
| | - Po-Lin Kuo
- Graduate Institute of Clinical Medicine, College of Medicine, Kaohsiung Medical University, Kaohsiung 807, Taiwan.
| |
Collapse
|
28
|
Nie Y, Zhang D, Jin Z, Li B, Wang X, Che H, You Y, Qian X, Zhang Y, Zhao P, Chai G. Lanatoside C protects mice against bleomycin-induced pulmonary fibrosis through suppression of fibroblast proliferation and differentiation. Clin Exp Pharmacol Physiol 2019; 46:575-586. [PMID: 30854687 DOI: 10.1111/1440-1681.13081] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2018] [Revised: 02/21/2019] [Accepted: 03/04/2019] [Indexed: 12/14/2022]
Abstract
It has been established that lanatoside C, a FDA-approved cardiac glycoside, reduces proliferation of cancer cell lines. The proliferation of fibroblasts is critical to the pathogenesis of pulmonary fibrosis (PF), a progressive and fatal fibrotic lung disease lacking effective treatment. In this study we have investigated the impact of lanatoside C on a bleomycin (BLM)-induced mouse model of PF and through the evaluation of fibroblast proliferation and activation in vitro. We evaluated explanted lung tissue by histological staining, western blot analysis, qRT-PCR and survival analysis, demonstrating that lanatoside C was able to protect mice against BLM-induced pulmonary fibrosis. The proliferation of cultured pulmonary fibroblasts isolated from BLM-induced PF mice was suppressed by lanatoside C, as hypothesized, through the induction of cell apoptosis and cell cycle arrest at the G2/M phase. The Akt signalling pathway was involved in this process. Interestingly, the production of α-SMA, fibronectin, and collagen I and III in response to TGF-β1 in healthy mouse fibroblasts was suppressed following lanatoside C administration by inhibition of TGF-β1/Smad signalling. In addition, TGF-β1-induced migration in lung fibroblasts was also impeded after lanatoside C treatment. Together, our data revealed that lanatoside C alleviated BLM-induced pulmonary fibrosis in mice via attenuation of growth and differentiation of fibroblasts, suggesting that it has potential as a candidate therapy for PF patients.
Collapse
Affiliation(s)
- Yunjuan Nie
- Department of Basic Medicine, Wuxi School of Medicine, Jiangnan University, Wuxi, Jiangsu, China
| | - Dan Zhang
- Department of Laboratory Medicine, Research Center for Cancer Precision Medicine, Bengbu Medical College, Bengbu, Anhui Province, China
| | - Zhewu Jin
- Department of Basic Medicine, Wuxi School of Medicine, Jiangnan University, Wuxi, Jiangsu, China
| | - Boyu Li
- Department of Basic Medicine, Wuxi School of Medicine, Jiangnan University, Wuxi, Jiangsu, China
| | - Xue Wang
- Department of Basic Medicine, Wuxi School of Medicine, Jiangnan University, Wuxi, Jiangsu, China
| | - Huilian Che
- Department of Basic Medicine, Wuxi School of Medicine, Jiangnan University, Wuxi, Jiangsu, China
| | - Yaqian You
- Department of Basic Medicine, Wuxi School of Medicine, Jiangnan University, Wuxi, Jiangsu, China
| | - Xiaohang Qian
- Department of Basic Medicine, Wuxi School of Medicine, Jiangnan University, Wuxi, Jiangsu, China
| | - Yang Zhang
- Department of Orthopedic, Lu'an Fourth People's Hospital, Lu'an, Anhui, China
| | - Peng Zhao
- Department of Basic Medicine, Wuxi School of Medicine, Jiangnan University, Wuxi, Jiangsu, China
| | - Gaoshang Chai
- Department of Basic Medicine, Wuxi School of Medicine, Jiangnan University, Wuxi, Jiangsu, China
| |
Collapse
|
29
|
Chen DQ, Feng YL, Cao G, Zhao YY. Natural Products as a Source for Antifibrosis Therapy. Trends Pharmacol Sci 2018; 39:937-952. [PMID: 30268571 DOI: 10.1016/j.tips.2018.09.002] [Citation(s) in RCA: 156] [Impact Index Per Article: 22.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/04/2018] [Revised: 08/29/2018] [Accepted: 09/04/2018] [Indexed: 01/15/2023]
Abstract
Although fibrosis is a final pathological feature of many chronic diseases, few interventions are available that specifically target the pathogenesis of fibrosis. Natural products are becoming increasingly recognized as effective therapies for fibrosis. The highlights of common cellular and molecular mechanisms of fibrosis facilitate the discovery of effective antifibrotic drugs. We describe some new profibrotic mechanisms and corresponding therapeutic targets using natural products. Interleukin, ephrin-B2, Gas6/TAM, Wnt/β-catenin, hedgehog pathway, PPARγ, lysophosphatidic acid, and CTGF are promising therapeutic targets. Natural products can target these mediators and inhibit chronic inflammation, myofibroblast activation, epithelial-mesenchymal transition, and extracellular matrix accumulation to alleviate fibrosis. Of note, natural products have the potential to inhibit fibrosis in one organ, simultaneously targeting fibrosis in multiple other organs, which provides us new strategies to find antifibrotic drugs.
Collapse
Affiliation(s)
- Dan-Qian Chen
- School of Life Science, Northwest University, No. 229 Taibai North Road, Xi'an, Shaanxi 710069, China
| | - Ya-Long Feng
- School of Life Science, Northwest University, No. 229 Taibai North Road, Xi'an, Shaanxi 710069, China
| | - Gang Cao
- School of Pharmacy, Zhejiang Chinese Medical University, No. 548 Binwen Road, Hangzhou, Zhejiang 310053, China
| | - Ying-Yong Zhao
- School of Life Science, Northwest University, No. 229 Taibai North Road, Xi'an, Shaanxi 710069, China.
| |
Collapse
|
30
|
Suppression of SMOC2 reduces bleomycin (BLM)-induced pulmonary fibrosis by inhibition of TGF-β1/SMADs pathway. Biomed Pharmacother 2018; 105:841-847. [DOI: 10.1016/j.biopha.2018.03.058] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2018] [Revised: 03/10/2018] [Accepted: 03/11/2018] [Indexed: 12/14/2022] Open
|
31
|
Kocak A, Harmancı D, Birlik M, Sarıoglu S, Yılmaz O, Cavdar Z, Guner G. Effects of epigallocatechin-3-gallate (EGCG) on a scleroderma model of fibrosis. TURKISH JOURNAL OF BIOCHEMISTRY 2018; 43:464-473. [DOI: 10.1515/tjb-2017-0185] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 07/01/2024]
Abstract
Abstract
Objective
The aim of the present study was to evaluate the potential protective effects of epigallocatechin-3-gallate (EGCG) on fibrosis in bleomycin induced scleroderma model.
Materials and methods
Thirty-two healthy female Balb-c mice with the average body weight of 22±5 g were used in this study. The mice were randomly divided into four groups as control (n=8), Bleomycin (n=8), Bleomycin+EGCG (n=8) and EGCG (n=8). Skin tissue samples were collected to quantify matrix metalloproteinases (MMP-1, MMP-8, MMP-13), p-SMAD 2/3 and SMAD 2/3 in protein homogenates by western blotting. TGF-β1 expression was determined by real-time PCR. Immunohistopathological and histopathological examinations of skin tissues were also done.
Results
When measured with Masson Trichrome, EGCG treatment was found to decrease fibrosis in connective tissue compared to the BLM injected control. EGCG was decreased dermal fibrosis. Bleomycin+EGCG group showed a significant reduction in fibrosis at the dermal surface area using hematoxylin measurements compared with the BLM group. MMP-1, MMP-8 protein levels were increased and p-SMAD 2/3 protein level was decreased. TGF-β mRNA expression was decreased in the EGCG+BLM group compared with the BLM group.
Conclusion
These results suggest an antifibrotic role for EGCG.
Collapse
Affiliation(s)
- Ayse Kocak
- Institute of Health Sciences , Department of Molecular Medicine , Dokuz Eylul University , Izmir , Turkey
| | - Duygu Harmancı
- Institute of Health Sciences , Department of Molecular Medicine , Dokuz Eylul University , Izmir , Turkey
| | - Merih Birlik
- Faculty of Medicine, Internal Medicine, Division of Rheumatology , Dokuz Eylul University , Izmir , Turkey
| | - Sulen Sarıoglu
- Faculty of Medicine, Department of Medical Pathology , Dokuz Eylül University , Izmir , Turkey
| | - Osman Yılmaz
- Institute of Health Sciences, Department of Laboratory Animal Sciences , Dokuz Eylul University , Izmir , Turkey
| | - Zahide Cavdar
- Institute of Health Sciences , Department of Molecular Medicine , Dokuz Eylul University , Izmir , Turkey
| | - Gul Guner
- Department of Biochemistry, Faculty of Medicine , Izmir University of Economics , Izmir , Turkey
| |
Collapse
|
32
|
Chen K, Chen W, Liu SL, Wu TS, Yu KF, Qi J, Wang Y, Yao H, Huang XY, Han Y, Hou P. Epigallocatechingallate attenuates myocardial injury in a mouse model of heart failure through TGF‑β1/Smad3 signaling pathway. Mol Med Rep 2018; 17:7652-7660. [PMID: 29620209 PMCID: PMC5983962 DOI: 10.3892/mmr.2018.8825] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2017] [Accepted: 12/15/2017] [Indexed: 12/19/2022] Open
Abstract
The present study aimed to assess the protective effect of epigallocatechingallate (EGCG) against myocardial injury in a mouse model of heart failure and to determine the mechanism underlying regulation of the transforming growth factor-β1/mothers against decapentaplegic homolog 3 (TGF-β1/Smad3) signaling pathway. Mouse models of heart failure were established. Alterations in ejection fraction, left ventricular internal diastolic diameter (LVIDd) and left ventricular internal systolic diameter (LVIDs) were measured by echocardiography. Pathological alterations of myocardial tissue were determined by hematoxylin and eosin, and Masson staining. The levels of serum brain natriuretic peptide (BNP), N-terminal-proBNP, interleukin (IL)-1β, IL-6, tumor necrosis factor-α, malondialdehyde, superoxide dismutase and glutathione peroxidase were detected with ELISA. Expression of collagen I, collagen III were detected by western blotting and reverse transcription quantitative polymerase chain reaction. Transforming growth factor-β1 (TGF-β1), Smad3, phosphorylated (p)-Smad3, apoptosis regulator BAX (Bax), caspase-3 and apoptosis regulator Bcl2 in mouse cardiac tissue were measured by western blotting. P-smad3 and TGF-β1 were measured by immunofluorescence staining. EGCG reversed the alterations in LVIDd and LVIDs induced by establishment of the model of heart failure, increased ejection fraction, inhibited myocardial fibrosis, attenuated the oxidative stress, inflammatory and cardiomyocyte apoptosis and lowered the expression levels of collagen I and collagen III. Following treatment with TGF-β1 inhibitor, the protective effect of EGCG against heart failure was attenuated. The results of the present study demonstrated that EGCG can inhibit the progression and development of heart failure in mice through inhibition of myocardial fibrosis and reduction of ventricular collagen remodeling. This protective effect of EGCG is likely mediated through inhibition of TGF-β1/smad3 signaling pathway.
Collapse
Affiliation(s)
- Keyan Chen
- Department of Laboratory Animal Science, China Medical University, Shenyang, Liaoning 110122, P.R. China
| | - Wei Chen
- Department of Cardiology, The First Clinical College, Liaoning University of Traditional Chinese Medicine, Shenyang, Liaoning 110032, P.R. China
| | - Shi Li Liu
- Department of Cardiology, The First Clinical College, Liaoning University of Traditional Chinese Medicine, Shenyang, Liaoning 110032, P.R. China
| | - Tian Shi Wu
- Department of Cardiology, The First Clinical College, Liaoning University of Traditional Chinese Medicine, Shenyang, Liaoning 110032, P.R. China
| | - Kai Feng Yu
- Department of Cardiology, The First Clinical College, Liaoning University of Traditional Chinese Medicine, Shenyang, Liaoning 110032, P.R. China
| | - Jing Qi
- Department of Cardiology, The First Clinical College, Liaoning University of Traditional Chinese Medicine, Shenyang, Liaoning 110032, P.R. China
| | - Yijun Wang
- Department of Cardiology, The First Clinical College, Liaoning University of Traditional Chinese Medicine, Shenyang, Liaoning 110032, P.R. China
| | - Hui Yao
- Department of Cardiology, The First Clinical College, Liaoning University of Traditional Chinese Medicine, Shenyang, Liaoning 110032, P.R. China
| | - Xiao Yang Huang
- Department of Cardiology, The First Clinical College, Liaoning University of Traditional Chinese Medicine, Shenyang, Liaoning 110032, P.R. China
| | - Ying Han
- Department of Cardiology, Jinqiu Hospital of Liaoning Province, Shenyang, Liaoning 110016, P.R. China
| | - Ping Hou
- Department of Cardiology, The First Clinical College, Liaoning University of Traditional Chinese Medicine, Shenyang, Liaoning 110032, P.R. China
| |
Collapse
|
33
|
Ciebiera M, Łukaszuk K, Męczekalski B, Ciebiera M, Wojtyła C, Słabuszewska-Jóźwiak A, Jakiel G. Alternative Oral Agents in Prophylaxis and Therapy of Uterine Fibroids-An Up-to-Date Review. Int J Mol Sci 2017; 18:ijms18122586. [PMID: 29194370 PMCID: PMC5751189 DOI: 10.3390/ijms18122586] [Citation(s) in RCA: 32] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2017] [Revised: 11/28/2017] [Accepted: 11/28/2017] [Indexed: 01/18/2023] Open
Abstract
Uterine fibroids (UFs) are the most common tumors of the female genital tract. The effect of UFs on the quality of life and the overall cost of treatment are significant issues worldwide. Tumor size and location are the two specific factors which influence the occurrence of symptoms, the need for, and method of, treatment (some tumors require surgery while some can be treated with selected drugs). Primary prevention and treatment of early UF disease are worthy goals that might have a great impact on health care systems. Several treatments and prophylactic methods can be used in this endeavor. This publication presents current data about lesser-known substances which may have a beneficial effect on the treatment or prophylaxis of UFs and can be administered orally, serving as an alternative to (or complement of) surgery or selective progesterone receptor modulators (SPRMs). Early prevention and treatment of UFs in women from high-risk groups should be our priority. Innovative forms of UF management are under intensive investigation and may be promising options in the near future. Many of them evaluated vitamin D, paricalcitol, epigallocatechin gallate (EGCG), elagolix, aromatase inhibitors (AIs), and cabergoline and deemed them to be safe and effective. The next step in such projects should be properly constructed randomized control trials (RCTs), carried out by successive phases.
Collapse
Affiliation(s)
- Michał Ciebiera
- Department of Obstetrics and Gynecology, The Centre of Postgraduate Medical Education, 00-416 Warsaw, Poland.
| | - Krzysztof Łukaszuk
- Department of Obstetrics and Gynecological Nursing, Faculty of Health Sciences, Medical University of Gdansk, 80-210 Gdansk, Poland.
- INVICTA Fertility and Reproductive Center, 80-172 Gdansk, Poland.
| | - Błażej Męczekalski
- Department of Gynecological Endocrinology, Poznan University of Medical Sciences, 60-513 Poznan, Poland.
| | - Magdalena Ciebiera
- Students' Scientific Association at the I Department of Obstetrics and Gynecology, Medical University of Warsaw, 02-015 Warsaw, Poland.
| | - Cezary Wojtyła
- Department of Obstetrics and Gynecology, The Centre of Postgraduate Medical Education, 00-416 Warsaw, Poland.
| | - Aneta Słabuszewska-Jóźwiak
- Department of Obstetrics and Gynecology, The Centre of Postgraduate Medical Education, 00-416 Warsaw, Poland.
| | - Grzegorz Jakiel
- Department of Obstetrics and Gynecology, The Centre of Postgraduate Medical Education, 00-416 Warsaw, Poland.
| |
Collapse
|
34
|
Guan S, Zhou J. Frizzled-7 mediates TGF-β-induced pulmonary fibrosis by transmitting non-canonical Wnt signaling. Exp Cell Res 2017; 359:226-234. [DOI: 10.1016/j.yexcr.2017.07.025] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2017] [Revised: 07/17/2017] [Accepted: 07/19/2017] [Indexed: 12/17/2022]
|
35
|
Wei Y, Kim TJ, Peng DH, Duan D, Gibbons DL, Yamauchi M, Jackson JR, Le Saux CJ, Calhoun C, Peters J, Derynck R, Backes BJ, Chapman HA. Fibroblast-specific inhibition of TGF-β1 signaling attenuates lung and tumor fibrosis. J Clin Invest 2017; 127:3675-3688. [PMID: 28872461 DOI: 10.1172/jci94624] [Citation(s) in RCA: 152] [Impact Index Per Article: 19.0] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2017] [Accepted: 07/18/2017] [Indexed: 12/12/2022] Open
Abstract
TGF-β1 signaling is a critical driver of collagen accumulation and fibrotic disease but also a vital suppressor of inflammation and epithelial cell proliferation. The nature of this multifunctional cytokine has limited the development of global TGF-β1 signaling inhibitors as therapeutic agents. We conducted phenotypic screens for small molecules that inhibit TGF-β1-induced epithelial-mesenchymal transition without immediate TGF-β1 receptor (TβR) kinase inhibition. We identified trihydroxyphenolic compounds as potent blockers of TGF-β1 responses (IC50 ~50 nM), Snail1 expression, and collagen deposition in vivo in models of pulmonary fibrosis and collagen-dependent lung cancer metastasis. Remarkably, the functional effects of trihydroxyphenolics required the presence of active lysyl oxidase-like 2 (LOXL2), thereby limiting effects to fibroblasts or cancer cells, the major LOXL2 producers. Mechanistic studies revealed that trihydroxyphenolics induce auto-oxidation of a LOXL2/3-specific lysine (K731) in a time-dependent reaction that irreversibly inhibits LOXL2 and converts the trihydrophenolic to a previously undescribed metabolite that directly inhibits TβRI kinase. Combined inhibition of LOXL2 and TβRI activities by trihydrophenolics resulted in potent blockade of pathological collagen accumulation in vivo without the toxicities associated with global inhibitors. These findings elucidate a therapeutic approach to attenuate fibrosis and the disease-promoting effects of tissue stiffness by specifically targeting TβRI kinase in LOXL2-expressing cells.
Collapse
Affiliation(s)
- Ying Wei
- Department of Medicine, UCSF Cardiovascular Research Institute, San Francisco, California, USA
| | - Thomas J Kim
- Department of Medicine, UCSF Cardiovascular Research Institute, San Francisco, California, USA
| | - David H Peng
- Departments of Thoracic/Head and Neck Medical Oncology and Molecular and Cellular Oncology, The University of Texas MD Anderson Cancer Center, Houston, Texas, USA
| | - Dana Duan
- Department of Cell and Tissue Biology, UCSF, San Francisco, California, USA
| | - Don L Gibbons
- Departments of Thoracic/Head and Neck Medical Oncology and Molecular and Cellular Oncology, The University of Texas MD Anderson Cancer Center, Houston, Texas, USA
| | - Mitsuo Yamauchi
- Oral and Craniofacial Health Sciences, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, USA
| | - Julia R Jackson
- Department of Medicine, UCSF Cardiovascular Research Institute, San Francisco, California, USA
| | - Claude J Le Saux
- Department of Medicine, UCSF Cardiovascular Research Institute, San Francisco, California, USA.,Department of Pulmonary and Critical Care, University of Texas Health Science Center at San Antonio, San Antonio, Texas, USA
| | - Cheresa Calhoun
- Department of Pulmonary and Critical Care, University of Texas Health Science Center at San Antonio, San Antonio, Texas, USA
| | - Jay Peters
- Department of Pulmonary and Critical Care, University of Texas Health Science Center at San Antonio, San Antonio, Texas, USA
| | - Rik Derynck
- Department of Cell and Tissue Biology, UCSF, San Francisco, California, USA
| | - Bradley J Backes
- Department of Medicine, UCSF Cardiovascular Research Institute, San Francisco, California, USA
| | - Harold A Chapman
- Department of Medicine, UCSF Cardiovascular Research Institute, San Francisco, California, USA
| |
Collapse
|
36
|
Li LC, Kan LD. Traditional Chinese medicine for pulmonary fibrosis therapy: Progress and future prospects. JOURNAL OF ETHNOPHARMACOLOGY 2017; 198:45-63. [PMID: 28038955 PMCID: PMC7127743 DOI: 10.1016/j.jep.2016.12.042] [Citation(s) in RCA: 98] [Impact Index Per Article: 12.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 09/12/2016] [Revised: 12/09/2016] [Accepted: 12/26/2016] [Indexed: 05/04/2023]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE Pulmonary fibrosis (PF) is a chronic, debilitating and often lethal lung disorder. Despite the molecular mechanisms of PF are gradually clear with numerous researchers' efforts, few effective drugs have been developed to reverse human PF or even halt the chronic progression to respiratory failure. Traditional Chinese medicine (TCM), the main component of the medical practice used for more than 5000 years especially in China, often exerts wider action spectrum than previously attempted options in treating human diseases. Recent data have shown the anti-fibrotic benefits of the active ingredients from TCM in this field, which may represent an attractive source of the drug discovery against PF. AIM OF THE REVIEW This review summarizes the pre-clinical and clinical evidence on the benefits of TCM and their active ingredients, and provides a comprehensive information and reliable basis for the exploration of new treatment strategies of botanical drugs in the therapy of PF. METHODS The literature information was obtained from the scientific databases on ethnobotany and ethno medicines (up to Aug 2016), mainly from the Pubmed, Web of Science and CNKI databases, and was to identify the experimental studies on the anti-fibrotic role of the active agents from TCM and the involved mechanisms. The search keywords for such work included: "lung fibrosis" or "pulmonary fibrosis", and "traditional Chinese medicine", "extract" or "herb". RESULTS A number of studies have shown that the active agents of single herbs and TCM formulas, particularly the flavonoids, glycosides and alkaloids, exhibit potential benefits against PF, the mechanisms of which appear to involve the regulation of inflammation, oxidant stress, and pro-fibrotic signaling pathways, etc. Besides, the processing methods for discovering TCM in treating PF were prospectively discussed. CONCLUSION These research work have shown the therapeutic benefits of TCM in the treatment of PF. However, more continued researches should be undertaken to clarify the unconfirmed chemical composition and regulatory mechanisms, conduct standard clinical trials, and evaluate the possible side effects. The insights provided in present review will be needed for further exploration of botanical drugs in the development of PF therapy.
Collapse
Affiliation(s)
- Liu-Cheng Li
- Department of Pharmacy, Sir Run Run Shaw Hospital, School of Medicine, Zhejiang University, Hangzhou 310016, China.
| | - Lian-Di Kan
- Department of Pharmacy, Sir Run Run Shaw Hospital, School of Medicine, Zhejiang University, Hangzhou 310016, China.
| |
Collapse
|
37
|
Guan S, Zhou J. CXCR7 attenuates the TGF-β-induced endothelial-to-mesenchymal transition and pulmonary fibrosis. MOLECULAR BIOSYSTEMS 2017; 13:2116-2124. [PMID: 28820530 DOI: 10.1039/c7mb00247e] [Citation(s) in RCA: 38] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
Lung fibrosis is a progressive and often fatal lung disease characterized by fibroblast proliferation and excessive deposition of extracellular matrix in the lungs.
Collapse
Affiliation(s)
- Shuhong Guan
- Department of Respiratory
- The First People's Hospital of Changzhou
- Changzhou 213003
- China
| | - Jun Zhou
- Department of Respiratory
- The First People's Hospital of Changzhou
- Changzhou 213003
- China
| |
Collapse
|
38
|
Epigallocatechin-3-Gallate Upregulates miR-221 to Inhibit Osteopontin-Dependent Hepatic Fibrosis. PLoS One 2016; 11:e0167435. [PMID: 27935974 PMCID: PMC5147893 DOI: 10.1371/journal.pone.0167435] [Citation(s) in RCA: 35] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2016] [Accepted: 11/14/2016] [Indexed: 02/06/2023] Open
Abstract
Osteopontin (OPN) promotes hepatic fibrosis, and developing therapies targeting OPN expression in settings of hepatic injury holds promise. The polyphenol epigallocatechin-3-gallate (EGCG), found in high concentrations in green tea, downregulates OPN expression through OPN mRNA degradation, but the mechanism is unknown. Previous work has shown that microRNAs can decrease OPN mRNA levels, and other studies have shown that EGCG modulates the expression of multiple microRNAs. In our study, we first demonstrated that OPN induces hepatic stellate cells to transform into an activated state. We then identified three microRNAs which target OPN mRNA: miR-181a, miR-10b, and miR-221. In vitro results show that EGCG upregulates all three microRNAs, and all three microRNAs are capable of down regulating OPN mRNA when administered alone. Interestingly, only miR-221 is necessary for EGCG-mediated OPN mRNA degradation and miR-221 inhibition reduces the effects of EGCG on cell function. In vivo experiments show that thioacetamide (TAA)-induced cell cytotoxicity upregulates OPN expression; treatment with EGCG blocks the effects of TAA. Furthermore, chronic treatment of EGCG in vivo upregulates all three microRNAs equally, suggesting that in more chronic treatment all three microRNAs are involved in modulating OPN expression. We conclude that in in vitro and in vivo models of TAA-induced hepatic fibrosis, EGCG inhibits OPN-dependent injury and fibrosis. EGCG works primarily by upregulating miR-221 to accelerate OPN degradation. EGCG may therefore have utility as a protective agent in settings of liver injury.
Collapse
|