51
|
Kim HH, Jeong SH, Park MY, Bhosale PB, Abusaliya A, Kim HW, Seong JK, Ahn M, Park KI, Heo JD, Kim YS, Kim GS. Potential Joint Protective and Anti-Inflammatory Effects of Integrin α vβ 3 in IL-1β-Treated Chondrocytes Cells. Biomedicines 2023; 11:2745. [PMID: 37893118 PMCID: PMC10603936 DOI: 10.3390/biomedicines11102745] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2023] [Revised: 10/06/2023] [Accepted: 10/09/2023] [Indexed: 10/29/2023] Open
Abstract
In osteoarthritis (OA), the articular cartilage covering the articular surface of the bone wears out, exposing the subchondral bone, and the synovial membrane surrounding the joint becomes inflamed, causing pain and deformity. OA causes pain, stiffness, and swelling, and discomfort in the knee when climbing stairs is a typical symptom. Although drug development studies are conducted to treat these inflammatory joint diseases, it is difficult to find conclusive research results which could reduce inflammation and slow cartilage tear. The development of drugs to relieve inflammatory pain often utilizes inflammatory triggers. Interleukins, one of the proteins in the limelight as pro-inflammatory factors, are immune-system-stimulating factors that promote the body's fight against harmful factors such as bacteria. In this study, inflammation was induced in Chondrocytes cells (Chon-001 cells) with IL-1β and then treated with integrin αvβ3 to show anti-inflammatory and chondrogenesis effects. Integrin αvβ3 was not toxic to Chon-001 cells in any concentration groups treated with or without IL-1β. COX-2 and iNOS, which are major markers of inflammation, were significantly reduced by integrin αvβ3 treatment. Expressions of p-ERK, p-JNK, and p-p38 corresponding to the MAPKs signaling pathway and p-IκBα and p-p65 corresponding to the NF-κB signaling pathway were also decreased in a dose-dependent manner upon integrin αvβ3 treatment, indicating that inflammation was inhibited, whereas treatment with integrin αvβ3 significantly increased the expression of ALP, RUNX2, BMP2, BMP4, Aggrecan, SOX9, and COL2A1, suggesting that osteogenesis and chondrogenesis were induced. These results suggest that integrin αvβ3 in-duces an anti-inflammatory effect, osteogenesis, and chondrogenesis on IL-1β-induced Chon-001 cells.
Collapse
Affiliation(s)
- Hun Hwan Kim
- Research Institute of Life Science and College of Veterinary Medicine, Gyeongsang National University, Jinju 52828, Republic of Korea; (H.H.K.); (S.H.J.); (M.Y.P.); (P.B.B.); (A.A.); (K.I.P.)
| | - Se Hyo Jeong
- Research Institute of Life Science and College of Veterinary Medicine, Gyeongsang National University, Jinju 52828, Republic of Korea; (H.H.K.); (S.H.J.); (M.Y.P.); (P.B.B.); (A.A.); (K.I.P.)
| | - Min Yeong Park
- Research Institute of Life Science and College of Veterinary Medicine, Gyeongsang National University, Jinju 52828, Republic of Korea; (H.H.K.); (S.H.J.); (M.Y.P.); (P.B.B.); (A.A.); (K.I.P.)
| | - Pritam Bhagwan Bhosale
- Research Institute of Life Science and College of Veterinary Medicine, Gyeongsang National University, Jinju 52828, Republic of Korea; (H.H.K.); (S.H.J.); (M.Y.P.); (P.B.B.); (A.A.); (K.I.P.)
| | - Abuyaseer Abusaliya
- Research Institute of Life Science and College of Veterinary Medicine, Gyeongsang National University, Jinju 52828, Republic of Korea; (H.H.K.); (S.H.J.); (M.Y.P.); (P.B.B.); (A.A.); (K.I.P.)
| | - Hyun Wook Kim
- Division of Animal Bioscience & Intergrated Biotechnology, Jinju 52725, Republic of Korea;
| | - Je Kyung Seong
- Laboratory of Developmental Biology and Genomics, BK21 PLUS Program for Creative Veterinary Science Research, Research Institute for Veterinary Science, College of Veterinary Medicine, Seoul National University, Seoul 08826, Republic of Korea;
| | - Meejung Ahn
- Department of Animal Science, College of Life Science, Sangji University, Wonju 26339, Republic of Korea;
| | - Kwang Il Park
- Research Institute of Life Science and College of Veterinary Medicine, Gyeongsang National University, Jinju 52828, Republic of Korea; (H.H.K.); (S.H.J.); (M.Y.P.); (P.B.B.); (A.A.); (K.I.P.)
| | - Jeong Doo Heo
- Biological Resources Research Group, Bioenvironmental Science and Toxicology Division, Korea Institute of Toxicology Gyeongnam Branch (KIT), Jinju 52834, Republic of Korea;
| | - Young Sil Kim
- T-Stem Co., Ltd., Changwon 51573, Republic of Korea;
| | - Gon Sup Kim
- Research Institute of Life Science and College of Veterinary Medicine, Gyeongsang National University, Jinju 52828, Republic of Korea; (H.H.K.); (S.H.J.); (M.Y.P.); (P.B.B.); (A.A.); (K.I.P.)
| |
Collapse
|
52
|
Asghariazar V, Kadkhodayi M, Sarailoo M, Jolfayi AG, Baradaran B. MicroRNA-143 as a potential tumor suppressor in cancer: An insight into molecular targets and signaling pathways. Pathol Res Pract 2023; 250:154792. [PMID: 37689002 DOI: 10.1016/j.prp.2023.154792] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/22/2023] [Revised: 08/25/2023] [Accepted: 09/02/2023] [Indexed: 09/11/2023]
Abstract
MicroRNAs (MiRNAs), which are highly conserved and small noncoding RNAs, negatively regulate gene expression and influence signaling pathways involved in essential biological activities, including cell proliferation, differentiation, apoptosis, and cell invasion. MiRNAs have received much attention in the past decade due to their significant roles in cancer development. In particular, microRNA-143 (miR-143) is recognized as a tumor suppressor and is downregulated in most cancers. However, it seems that miR-143 is upregulated in rare cases, such as prostate cancer stem cells, and acts as an oncogene. The present review will outline the current studies illustrating the impact of miR-143 expression levels on cancer progression and discuss its target genes and their relevant signaling pathways to discover a potential therapeutic way for cancer.
Collapse
Affiliation(s)
- Vahid Asghariazar
- Cancer Immunology and Immunotherapy Research Center, Ardabil University of Medical Sciences, Ardabil, Iran; Immunology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran; Deputy of Research and Technology, Ardabil University of Medical Sciences, Ardabil, Iran.
| | - Mahtab Kadkhodayi
- Immunology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran; Department of Animal Biology, Faculty of Natural Sciences, The University of Tabriz, Tabriz, Iran
| | - Mehdi Sarailoo
- Students Research Committee, School of Medicine, Ardabil University of Medical Sciences, Ardabil, Iran
| | - Amir Ghaffari Jolfayi
- Student Research Committee, School of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Behzad Baradaran
- Immunology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran.
| |
Collapse
|
53
|
Delgado-Gonzalez P, Garza-Treviño EN, de la Garza Kalife DA, Quiroz Reyes A, Hernández-Tobías EA. Bioactive Compounds of Dietary Origin and Their Influence on Colorectal Cancer as Chemoprevention. Life (Basel) 2023; 13:1977. [PMID: 37895359 PMCID: PMC10608661 DOI: 10.3390/life13101977] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2023] [Revised: 09/22/2023] [Accepted: 09/25/2023] [Indexed: 10/29/2023] Open
Abstract
Colorectal cancer (CRC) is one of the most common causes of death and the third most diagnosed cancer worldwide. The tumor microenvironment and cancer stem cells participate in colorectal tumor progression and can dictate malignancy. Nutrition status affects treatment response and the progression or recurrence of the tumor. This review summarizes the main bioactive compounds against the molecular pathways related to colorectal carcinogenesis. Moreover, we focus on the compounds with chemopreventive properties, mainly polyphenols and carotenoids, which are highly studied dietary bioactive compounds present in major types of food, like vegetables, fruits, and seeds. Their proprieties are antioxidant and gut microbiota modulation, important in the intestine because they decrease reactive oxygen species and inflammation, both principal causes of cancer. These compounds can promote apoptosis and inhibit cell growth, proliferation, and migration. Combined with oncologic treatment, a sensitization to first-line colorectal chemotherapy schemes, such as FOLFOX and FOLFIRI, is observed, making them an attractive and natural support in the oncologic treatment of CRC.
Collapse
Affiliation(s)
- Paulina Delgado-Gonzalez
- Departamento de Bioquímica y Medicina Molecular, Facultad de Medicina, Universidad Autónoma de Nuevo León (UANL), Monterrey 6440, Mexico; (E.N.G.-T.); (D.A.d.l.G.K.); (A.Q.R.)
| | - Elsa N. Garza-Treviño
- Departamento de Bioquímica y Medicina Molecular, Facultad de Medicina, Universidad Autónoma de Nuevo León (UANL), Monterrey 6440, Mexico; (E.N.G.-T.); (D.A.d.l.G.K.); (A.Q.R.)
| | - David A. de la Garza Kalife
- Departamento de Bioquímica y Medicina Molecular, Facultad de Medicina, Universidad Autónoma de Nuevo León (UANL), Monterrey 6440, Mexico; (E.N.G.-T.); (D.A.d.l.G.K.); (A.Q.R.)
| | - Adriana Quiroz Reyes
- Departamento de Bioquímica y Medicina Molecular, Facultad de Medicina, Universidad Autónoma de Nuevo León (UANL), Monterrey 6440, Mexico; (E.N.G.-T.); (D.A.d.l.G.K.); (A.Q.R.)
| | | |
Collapse
|
54
|
Wang H, Wang Z, Zhang Z, Liu J, Hong L. β-Sitosterol as a Promising Anticancer Agent for Chemoprevention and Chemotherapy: Mechanisms of Action and Future Prospects. Adv Nutr 2023; 14:1085-1110. [PMID: 37247842 PMCID: PMC10509430 DOI: 10.1016/j.advnut.2023.05.013] [Citation(s) in RCA: 32] [Impact Index Per Article: 16.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2023] [Revised: 05/15/2023] [Accepted: 05/23/2023] [Indexed: 05/31/2023] Open
Abstract
Cancer is one of the primary causes of death worldwide, and its incidence continues to increase yearly. Despite significant advances in research, the search for effective and nontoxic preventive and therapeutic agents remains greatly important. Cancer is a multimodal disease, where various mechanisms play significant roles in its occurrence and progression. This highlights the need for multitargeted approaches that are not only safe and inexpensive but also provide effective alternatives for current therapeutic regimens. β-Sitosterol (SIT), the most abundant phytosterol found in various plant foods, represents such an option. Preclinical evidence over the past few decades has overwhelmingly shown that SIT exhibits multiple anticancer activities against varied cancers, such as liver, cervical, colon, stomach, breast, lung, pancreatic, and prostate cancers, in addition to leukemia, multiple myeloma, melanoma, and fibrosarcoma. In this article, we present the latest advances and perspectives on SIT-systematically summarizing its antitumor mechanisms of action into 7 main sections and combining current challenges and prospects-for its use as a promising agent for cancer prevention and treatment. In particular, SIT plays a role in cancer prevention and treatment mainly by enhancing apoptosis, inducing cell cycle arrest, bidirectionally regulating oxidative stress, improving metabolic reprogramming, inhibiting invasion and metastasis, modulating immunity and inflammation, and combating drug resistance. Although SIT holds such great promise, the poor aqueous solubility and bioavailability coupled with low targeting efficacy limit its therapeutic efficacy and clinical application. Further research on novel drug delivery systems may improve these deficiencies. Overall, through complex and pleiotropic mechanisms, SIT has good potential for tumor chemoprevention and chemotherapy. However, no clinical trials have yet proven this potential. This review provides theoretical basis and rationality for the further design and conduct of clinical trials to confirm the anticancer activity of SIT.
Collapse
Affiliation(s)
- Haoyu Wang
- Department of Obstetrics and Gynecology, Renmin Hospital of Wuhan University, Wuhan, China
| | - Zhi Wang
- Department of Obstetrics and Gynecology, Renmin Hospital of Wuhan University, Wuhan, China
| | - Zihui Zhang
- Department of Obstetrics and Gynecology, Renmin Hospital of Wuhan University, Wuhan, China
| | - Jingchun Liu
- Department of Obstetrics and Gynecology, Renmin Hospital of Wuhan University, Wuhan, China
| | - Li Hong
- Department of Obstetrics and Gynecology, Renmin Hospital of Wuhan University, Wuhan, China.
| |
Collapse
|
55
|
Khedr SM, Ghareeb DA, Fathy SA, Hamdy GM. Berberine-loaded albumin nanoparticles reverse aflatoxin B1-induced liver hyperplasia. BMC Pharmacol Toxicol 2023; 24:42. [PMID: 37559065 PMCID: PMC10413506 DOI: 10.1186/s40360-023-00683-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2022] [Accepted: 08/01/2023] [Indexed: 08/11/2023] Open
Abstract
Hepatocellular carcinoma (HCC) can be produced from aflatoxin B1 (AFB1) administration. Although berberine (BER) acts as an anticancer agent and can counteract the AFB1 effect, it has low bioavailability. Nanotechnology can overcome this problem. This research aimed to synthesize berberine nanoparticles (NPs) and then estimate their therapeutic effect compared to that of berberine against aflatoxin-induced hepatotoxicity. The desolvation method was used to prepare BER-NPs. Aflatoxicosis was induced by 5 consecutive intraperitoneal injections (IP) of 200 µg/kg/day AFB dissolved in dimethylsulfoxide (DMSO). After the induction period, two treatments were performed: the first with 100 mg/kg BER and the second with 10 mg/kg BER-NPs. Liver, kidney, and diabetic profiles were estimated by using standardized methods. Hepatic oxidative stress, inflammatory, cancer cell proliferation, and invasion markers were used by ELISA and qPCR techniques. The TEM image shows that both BSA NPs and BER-BSA NPs had spherical, regular, and uniform shapes. The BER encapsulation efficiency % was 78.5. The formed-BER-BSA NPs showed a loading capacity % of 7.71 and the synthesis yield % of 92.6. AFB1 increases pro-oxidant markers, decreases antioxidant systems, stimulates inflammatory enzymes, inhibits anti-inflammatory markers, decreases tumor suppressor enzymes, increases oncogenes, increases glycolytic activity, prevents cell death, and promotes cell growth. Most of the biochemical markers and hepatic architecture were normalized in the BER-BSA NP-treated group but not in the BER-treated group. Altogether, the obtained data proved that treatment with BER-NPs was more efficient than treatment with berberine against aflatoxicoses induced in rats.
Collapse
Affiliation(s)
- Sarah M Khedr
- Department of Biochemistry, Faculty of Science, Ain Shams University, Cairo, 11566, Egypt
| | - Doaa A Ghareeb
- Bio-Screening and Preclinical Trial Lab, Department of Biochemistry, Faculty of Science, Alexandria University, Alexandria, Egypt.
| | - Shadia A Fathy
- Department of Biochemistry, Faculty of Science, Ain Shams University, Cairo, 11566, Egypt
| | - Germine M Hamdy
- Department of Biochemistry, Faculty of Science, Ain Shams University, Cairo, 11566, Egypt
| |
Collapse
|
56
|
Chen D, Chen Y, Huang F, Zhang X, Zhou Y, Xu L. The underlying regulatory mechanisms of colorectal carcinoma by combining Vitexin and Aspirin: based on systems biology, molecular docking, molecular dynamics simulation, and in vitro study. Front Endocrinol (Lausanne) 2023; 14:1147132. [PMID: 37564983 PMCID: PMC10410442 DOI: 10.3389/fendo.2023.1147132] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/18/2023] [Accepted: 07/03/2023] [Indexed: 08/12/2023] Open
Abstract
Introduction Colorectal cancer (CRC) is a highly prevalent digestive system malignancy. Aspirin is currently one of the most promising chemopreventive agents for CRC, and the combination of aspirin and natural compounds helps to enhance the anticancer activity of aspirin. Natural flavonoids like vitexin have an anticancer activity focusing on colorectal carcinoma. Methods This study investigated the potential mechanism of action of the novel combination of vitexin and aspirin against colorectal cancer through network pharmacology, molecular docking, molecular dynamics simulation, and in vitro experiments. Results The results of network pharmacology suggested that vitexin and aspirin regulate multiple signaling pathways through various target proteins such as NFKB1, PTGS2 (COX-2), MAPK1, MAPK3, and TP53. Cellular experiments revealed that the combined effect of vitexin and aspirin significantly inhibited HT-29 cell growth. Vitexin dose-dependently inhibited COX-2 expression in cells and enhanced the down-regulation of COX-2 and NF-κB expression in colorectal cancer cells by aspirin. Discussion This study provides a pharmacodynamic material and theoretical basis for applying agents against colorectal cancer to delay the development of drug resistance and improve the prognosis of cancer patients.
Collapse
Affiliation(s)
- Dengsheng Chen
- Department of Clinical Pharmacy, Sanming First Hospital, Affiliated Hospital of Fujian Medical University, Sanming, Fujian, China
| | - Ying Chen
- Department of Clinical Pharmacy, Sanming First Hospital, Affiliated Hospital of Fujian Medical University, Sanming, Fujian, China
| | - Fang Huang
- Department of Clinical Pharmacy, Sanming First Hospital, Affiliated Hospital of Fujian Medical University, Sanming, Fujian, China
| | - Xiaoling Zhang
- Department of Clinical Pharmacy, Sanming First Hospital, Affiliated Hospital of Fujian Medical University, Sanming, Fujian, China
| | - Yulv Zhou
- Department of Chinese Medicine and Anorectology, Sanming First Hospital, Affiliated Hospital of Fujian Medical University, Sanming, Fujian, China
| | - Luning Xu
- Department of Clinical Pharmacy, Sanming First Hospital, Affiliated Hospital of Fujian Medical University, Sanming, Fujian, China
| |
Collapse
|
57
|
Fonseca Hernández D, Mojica L, Berhow MA, Brownstein K, Lugo Cervantes E, Gonzalez de Mejia E. Black and pinto beans (Phaseolus vulgaris L.) unique mexican varieties exhibit antioxidant and anti-inflammatory potential. Food Res Int 2023; 169:112816. [PMID: 37254392 DOI: 10.1016/j.foodres.2023.112816] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2022] [Revised: 03/14/2023] [Accepted: 04/11/2023] [Indexed: 06/01/2023]
Abstract
Oxidative stress and inflammation play a key role in diverse pathological conditions such as cancer and metabolic disorders. The objective of this study was to determine the antioxidant and anti-inflammatory potentials of crude extract (CE) and phenolic-enriched extract (PHE) obtained from the seed coats (SCs) of black bean (BB) and pinto bean (PB) varieties. Delphinidin-3-O-glucoside (46 mg/g SC), malvidin-3-O-glucoside (29.9 mg/g SC), and petunidin-3-O-glucoside (7.5 mg/g SC) were found in major concentrations in the PHE-BB. Pelargonidin (0.53 mg/g SC) was only identified in the PHE-PB. PHE from both varieties showed antioxidant and radical scavenging capacities, with strong correlations associated with total phenolic content (TPC). Polyphenolics, including catechin, myricetin, kaempferol, quercetin, and isorhamnetin glucosides, were identified in the extracts. In terms of the anti-inflammatory potentials, PHE-PB had an IC50 of 10.5 µg dry extract/mL (µg DE/mL) for cyclooxygenase-2 (COX-2) inhibition. The inhibition values for cyclooxygenase-1 (COX-1) ranged from 118.1 to 162.7 µg DE/mL. Regarding inducible nitric oxide synthase (iNOS) inhibition, PHE-BB had an IC50 of 62.6 µg DE/mL. As determined via in silico analysis, pelargonidin showed binding affinities of -7.8 and -8.5 kcal/mol for COX-1 and iNOS, respectively, and catechin had a value of -8.3 kcal/mol for COX-2. Phenolic-enriched extracts from seed coats of black and pinto beans showed good antioxidant and anti-inflammatory potential that warrants in vitro and in vivo studies.
Collapse
Affiliation(s)
- David Fonseca Hernández
- Tecnología Alimentaria, Centro de Investigación y Asistencia en Tecnología y Diseño del Estado de Jalisco, A.C., Guadalajara 44270, Mexico; Department of Food Science and Human Nutrition, University of Illinois at Urbana-Champaign, Champaign, IL 61801, USA.
| | - Luis Mojica
- Tecnología Alimentaria, Centro de Investigación y Asistencia en Tecnología y Diseño del Estado de Jalisco, A.C., Guadalajara 44270, Mexico.
| | - Mark A Berhow
- USDA, Agricultural Research Service, National Center for Agricultural Utilization Research, Function Food Research, 1815 N University, Peoria, IL 61604, USA
| | | | - Eugenia Lugo Cervantes
- Tecnología Alimentaria, Centro de Investigación y Asistencia en Tecnología y Diseño del Estado de Jalisco, A.C., Guadalajara 44270, Mexico
| | - Elvira Gonzalez de Mejia
- Department of Food Science and Human Nutrition, University of Illinois at Urbana-Champaign, Champaign, IL 61801, USA.
| |
Collapse
|
58
|
Li J, Chen J, Huang P, Cai Z, Zhang N, Wang Y, Li Y. The Anti-Inflammatory Mechanism of Flaxseed Linusorbs on Lipopolysaccharide-Induced RAW 264.7 Macrophages by Modulating TLR4/NF-κB/MAPK Pathway. Foods 2023; 12:2398. [PMID: 37372610 DOI: 10.3390/foods12122398] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2023] [Revised: 06/05/2023] [Accepted: 06/15/2023] [Indexed: 06/29/2023] Open
Abstract
Flaxseed linusorbs (FLs), cyclic peptides derived from flaxseed oils, have shown multiple activities such as anticancer, antibacterial, and anti-inflammatory effects. However, the anti-inflammatory monomers of FLs and their mechanisms are still unclear. In this study, we have elucidated that FLs suppress the modulation of NF-κB/MAPK signaling pathways by targeting the inhibition of activating TLR4 in LPS-induced RAW 264.7 cells. Therefore, the transcription and expression of inflammatory cytokines (i.e., TNF-α, IL-1β, and IL-6) and inflammatory mediator proteins (i.e., iNos and Cox-2) were significantly suppressed by FLs. In addition, an in silico study discovered that eight monomers of FLs showed high-affinity bindings with TLR4. In silico data combined with HPLC results indicated that FLA and FLE, accounting for 44%, were likely the major anti-inflammatory monomers in FLs. In summary, FLA and FLE were proposed as the main anti-inflammatory active cyclopeptides via hindering TLR4/NF-κB/MAPK signaling pathways, suggesting the potential use of food-derived FLs as natural anti-inflammatory supplements in a daily diet.
Collapse
Affiliation(s)
- Jialong Li
- Guangdong International Joint Research Center for Oilseeds Biorefinery, Nutrition and Safety, Department of Food Science and Engineering, Jinan University, Guangzhou 510632, China
| | - Jing Chen
- Guangdong International Joint Research Center for Oilseeds Biorefinery, Nutrition and Safety, Department of Food Science and Engineering, Jinan University, Guangzhou 510632, China
- Institute for Advance and Application Chemical Synthesis, Jinan University, Guangzhou 510632, China
| | - Ping Huang
- Guangzhou Meizhiao Cosmetics Co., Ltd., No. 555, Panyu Av. North, Guangzhou 510000, China
| | - Zizhe Cai
- Guangdong International Joint Research Center for Oilseeds Biorefinery, Nutrition and Safety, Department of Food Science and Engineering, Jinan University, Guangzhou 510632, China
| | - Ning Zhang
- Guangdong International Joint Research Center for Oilseeds Biorefinery, Nutrition and Safety, Department of Food Science and Engineering, Jinan University, Guangzhou 510632, China
| | - Yong Wang
- Guangdong International Joint Research Center for Oilseeds Biorefinery, Nutrition and Safety, Department of Food Science and Engineering, Jinan University, Guangzhou 510632, China
| | - Ying Li
- Guangdong International Joint Research Center for Oilseeds Biorefinery, Nutrition and Safety, Department of Food Science and Engineering, Jinan University, Guangzhou 510632, China
| |
Collapse
|
59
|
Cai H, Chen Y, Zhang Q, Liu Y, Jia H. High preoperative CEA and systemic inflammation response index (C-SIRI) predict unfavorable survival of resectable colorectal cancer. World J Surg Oncol 2023; 21:178. [PMID: 37291634 DOI: 10.1186/s12957-023-03056-z] [Citation(s) in RCA: 16] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2023] [Accepted: 05/28/2023] [Indexed: 06/10/2023] Open
Abstract
BACKGROUND CEA and systemic inflammation were reported to correlate with proliferation, invasion, and metastasis of colorectal cancer. This study investigated the prognostic significance of the preoperative CEA and systemic inflammation response index (C-SIRI) in patients with resectable colorectal cancer. METHODS Two hundred seventeen CRC patients were recruited from Chongqing Medical University, the first affiliated hospital, between January 2015 and December 2017. Baseline characteristics, preoperative CEA level, and peripheral monocyte, neutrophil, and lymphocyte counts were retrospectively reviewed. The optimal cutoff value for SIRI was defined as 1.1, and for CEA, the best cutoff values were 4.1 ng/l and 13.0 ng/l. Patients with low levels of CEA (< 4.1 ng/l) and SIRI (< 1.1) were assigned a value of 0, those with high levels of CEA (≥ 13.0 ng/l) and SIRI (≥ 1.1) were assigned a value of 3, and those with CEA (4.1-13.0 ng/l) and SIRI (≥ 1.1), CEA (≥ 13.0 ng/l), and SIRI (< 1.1) were assigned a value of 2. Those with CEA (< 4.1 ng/l) and SIRI (≥ 1.1) and CEA (4.1-13.0 ng/l) and SIRI (< 1.1) were assigned a value of 1. The prognostic value was assessed based on univariate and multivariate survival analysis. RESULTS Preoperative C-SIRI was statistically correlated with gender, site, stage, CEA, OPNI, NLR, PLR, and MLR. However, no difference was observed between C-SIRI and age, BMI, family history of cancer, adjuvant therapy, and AGR groups. Among these indicators, the correlation between PLR and NLR is the strongest. In addition, high preoperative C-SIRI was significantly correlated with poorer overall survival (OS) (HR: 2.782, 95% CI: 1.630-4.746, P < 0.001) based on univariate survival analysis. Moreover, it remained an independent predictor for OS (HR: 2.563, 95% CI: 1.419-4.628, p = 0.002) in multivariate Cox regression analysis. CONCLUSION Our study showed that preoperative C-SIRI could serve as a significant prognostic biomarker in patients with resectable colorectal cancer.
Collapse
Affiliation(s)
- Hao Cai
- Department of Gastrointestinal Surgery, The First Affiliated Hospital of Chongqing Medical University, No.1, Medical College Road, Yuzhong District, Chongqing, 400016, China
| | - Yu Chen
- Department of Gastrointestinal Surgery, The First Affiliated Hospital of Chongqing Medical University, No.1, Medical College Road, Yuzhong District, Chongqing, 400016, China
| | - Qiao Zhang
- Department of Gastrointestinal Surgery, The First Affiliated Hospital of Chongqing Medical University, No.1, Medical College Road, Yuzhong District, Chongqing, 400016, China
| | - Yang Liu
- Department of Gastrointestinal Surgery, The First Affiliated Hospital of Chongqing Medical University, No.1, Medical College Road, Yuzhong District, Chongqing, 400016, China
| | - HouJun Jia
- Department of Gastrointestinal Surgery, The First Affiliated Hospital of Chongqing Medical University, No.1, Medical College Road, Yuzhong District, Chongqing, 400016, China.
| |
Collapse
|
60
|
Sayed HM, Ramadan MA, Salem HH, Ahmad I, Patel H, Fayed MAA. Phytochemical Investigation, In Silico/In Vivo Analgesic, and Anti-inflammatory Assessment of the Egyptian Cassia occidentalis L. Steroids 2023; 196:109245. [PMID: 37141980 DOI: 10.1016/j.steroids.2023.109245] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/22/2022] [Revised: 04/24/2023] [Accepted: 04/26/2023] [Indexed: 05/06/2023]
Abstract
Cassia occidentalis L., from Fabaceae family phytochemical screening, revealed several biologically active principles mainly flavonoids and anthraquinones. GLC analysis of the lipoidal matter afforded 12 hydrocarbons: 9-dodecyl-tetradecahydro-anthracene (48.97 %), 9-dodecyl-tetradecahydro-phenanthrene (14.43 %), and 6 sterols/triterpenes: iso-jaspisterol (11.99%) and fatty acids were palmitic acid (50 %), and Linoleic acid (16.06%). Column chromatography led to the isolation of fifteen compounds (1-15), elucidated using spectroscopic evidence. First report of undecanoic acid (4) from the family Fabaceae, while p-dimethyl amino-benzaldehyde (15) was first time isolated from a natural origin. Eight compounds isolated for the first time from C. occidentalis L.; β-amyrin (1), β-sitosterol (2), stigmasterol (3), camphor (5), lupeol (6), chrysin (7), pectolinargenin (8), and 1, 2, 5-trihydroxy anthraquinone (14) besides five known compounds previously isolated; apigenin (9), kaempferol (10), chrysophanol (11), physcion (12), and aloe-emodin (13). In-vivo evaluation of anti-inflammatory and analgesic effects of C. occidentalis L. extracts where the n-butanol and total extracts showed the highest activities. The percentage of the inhibitory effect of the n-butanol extract was 29.7 at a dose of 400 mg/Kg. Furthermore, identified phytoconstituents were docked into the active sites of enzymes nAChRs, COX-1, and COX-2 to evaluate binding affinity. Phyto-compounds Physcion, aloe-emodin, and chrysophanol were found to have a good affinity for targeted receptors compared to co-crystalized inhibitors, validating the analgesic and anti-inflammatory effects of the phytochemicals.
Collapse
Affiliation(s)
- Hanaa M Sayed
- Department of Pharmacognosy, Faculty of Pharmacy, Assiut University, Assiut 71526, Egypt
| | - Mahmoud A Ramadan
- Department of Pharmacognosy, Faculty of Pharmacy, Assiut University, Assiut 71526, Egypt
| | - Heba H Salem
- Department of Pharmacognosy, Faculty of Pharmacy, Northern Border University, Rafha Region, Saudi Arabia
| | - Iqrar Ahmad
- Division of Computer Aided Drug Design, Department of Pharmaceutical Chemistry, R. C. Patel Institute of Pharmaceutical Education and Research, Shirpur,425405, Maharashtra, India
| | - Harun Patel
- Division of Computer Aided Drug Design, Department of Pharmaceutical Chemistry, R. C. Patel Institute of Pharmaceutical Education and Research, Shirpur,425405, Maharashtra, India
| | - Marwa A A Fayed
- Department of Pharmacognosy, Faculty of Pharmacy, University of Sadat City, Sadat City 32897, Egypt.
| |
Collapse
|
61
|
Kang S, Son Y, Shin IS, Moon C, Lee MY, Lim KS, Park SJ, Lee CG, Jo WS, Lee HJ, Kim JS. EFFECT OF ABDOMINAL IRRADIATION IN MICE MODEL OF INFLAMMATORY BOWEL DISEASE. RADIATION PROTECTION DOSIMETRY 2023; 199:564-571. [PMID: 36917812 DOI: 10.1093/rpd/ncad051] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/16/2022] [Revised: 02/01/2023] [Accepted: 02/02/2023] [Indexed: 06/18/2023]
Abstract
Inflammatory bowel diseases could be diagnosed in major measure by diagnostic imaging; however, radiation exposure in the intestine may also contribute to the progression of these pathologies. To better understand the impact of radiation in the presence of bowel disease, we administered dextran sodium sulfate (DSS) to C57BL/6 mice to induce colitis and exposed to radiation at abdominal area. We observed that abdominal irradiation (13 Gy) aggravates the DSS-induced decrease in survival rate (0%), body weight (74.54 ± 3.59%) and colon length (4.98 ± 0.14 cm). Additionally, abdominal irradiation markedly increased in colonic inflammation levels (3.16 ± 0.16) compared with that of DSS-induced sham mice. Furthermore, abdominal irradiation also increased the mRNA expression levels of inflammatory genes, such as cyclooxygenase-2 (13.10 folds), interleukin-6 (48.83 folds) and tumor necrosis factor-alpha (42.97 folds). We conclude that abdominal irradiation aggravates the detrimental effects of DSS-induced colitis in mice, which might be a useful guideline for inflammatory bowel disease patients.
Collapse
Affiliation(s)
- Sohi Kang
- College of Veterinary Medicine and BK21 FOUR Program, Chonnam National University, Gwangju 61186, The Republic of Korea
| | - Yeonghoon Son
- Division of Radiation Biomedical Research, Korea Institute of Radiological & Medical Sciences, Seoul 01812, The Republic of Korea
| | - In-Sik Shin
- College of Veterinary Medicine and BK21 FOUR Program, Chonnam National University, Gwangju 61186, The Republic of Korea
| | - Changjong Moon
- College of Veterinary Medicine and BK21 FOUR Program, Chonnam National University, Gwangju 61186, The Republic of Korea
| | - Min Y Lee
- College of Pharmacy, Research Institute of Pharmaceutical Sciences, Kyungpook National University, Daegu 41566, The Republic of Korea
| | - Kyung S Lim
- Futuristic Animal Resource and Research Center, Korea Research Institute of Bioscience and Biotechnology, Ochang 28116, Chungbuk, The Republic of Korea
| | - Su-Jin Park
- Futuristic Animal Resource and Research Center, Korea Research Institute of Bioscience and Biotechnology, Ochang 28116, Chungbuk, The Republic of Korea
| | - Chang-Geun Lee
- Research Center, Dongnam Institute of Radiological & Medical Sciences, Busan 46033, The Republic of Korea
| | - Wol S Jo
- Research Center, Dongnam Institute of Radiological & Medical Sciences, Busan 46033, The Republic of Korea
| | - Hae-June Lee
- Division of Radiation Biomedical Research, Korea Institute of Radiological & Medical Sciences, Seoul 01812, The Republic of Korea
| | - Joong S Kim
- College of Veterinary Medicine and BK21 FOUR Program, Chonnam National University, Gwangju 61186, The Republic of Korea
| |
Collapse
|
62
|
Lan K, Yang H, Zheng J, Hu H, Zhu T, Zou X, Hu B, Liu H. Poria cocos oligosaccharides ameliorate dextran sodium sulfate-induced colitis mice by regulating gut microbiota dysbiosis. Food Funct 2023; 14:857-873. [PMID: 36537246 DOI: 10.1039/d2fo03424g] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
Poria cocos, a widely accepted function food in China, has multiple pharmacological activities. This study aimed to investigate the therapeutic effect and molecular mechanism of Poria cocos oligosaccharides (PCOs) against dextran sodium sulfate (DSS)-induced mouse colitis. In this study, BALB/c mice were treated with 3% (w/v) DSS for seven days to establish a colitis model. The results showed that oral administration of PCOs (200 mg per kg per day) significantly reversed the changes in the physiological indices in colitis mice, including body weight, disease activity index scores (DAI), spleen index, and colon length. From the qRT-PCR assay, it was observed that PCOs suppressed the mRNA expression of pro-inflammatory cytokines, such as Tnf-α, Il-1β, and Il-6. In addition, PCOs protected the intestinal barrier from damage by promoting the expression of mucins and tight junction proteins at both mRNA and protein levels. Upon 16S rDNA sequencing, it was observed that PCO treatment partly reversed the changes in the gut microbiota of colitis mice by selectively regulating the abundance of specific bacteria. And Odoribacter, Muribaculum, Desulfovibrio, Oscillibacter, Escherichia-Shigella, and Turicibacter might be the critical bacteria in improving colitis via PCOs. Finally, using antibiotic mixtures to destroy the intestinal bacteria, we documented that PCO fermentation broth (PCO FB) instead of PCOs prevented the occurrence of colitis in gut microbiota-depleted mice. In conclusion, PCOs showed a protective effect on colitis by reversing gut microbiota dysbiosis. Our study sheds light on the potential application of PCOs as a prebiotic for treating colitis.
Collapse
Affiliation(s)
- Ke Lan
- College of Basic Medical Sciences, Hubei University of Chinese Medicine, Huangjiahu West Road 16, Wuhan 430065, P. R. China.
| | - Huabing Yang
- College of Basic Medical Sciences, Hubei University of Chinese Medicine, Huangjiahu West Road 16, Wuhan 430065, P. R. China.
| | - Junping Zheng
- College of Basic Medical Sciences, Hubei University of Chinese Medicine, Huangjiahu West Road 16, Wuhan 430065, P. R. China.
| | - Haiming Hu
- College of Basic Medical Sciences, Hubei University of Chinese Medicine, Huangjiahu West Road 16, Wuhan 430065, P. R. China.
| | - Tianxiang Zhu
- College of Basic Medical Sciences, Hubei University of Chinese Medicine, Huangjiahu West Road 16, Wuhan 430065, P. R. China.
| | - Xiaojuan Zou
- College of Basic Medical Sciences, Hubei University of Chinese Medicine, Huangjiahu West Road 16, Wuhan 430065, P. R. China.
| | - Baifei Hu
- College of Basic Medical Sciences, Hubei University of Chinese Medicine, Huangjiahu West Road 16, Wuhan 430065, P. R. China.
| | - Hongtao Liu
- College of Basic Medical Sciences, Hubei University of Chinese Medicine, Huangjiahu West Road 16, Wuhan 430065, P. R. China.
| |
Collapse
|
63
|
Mola-Ali-Nejad R, Fakharianzadeh S, Maloum Z, Taheri M, Shirvani-Farsani Z. A gene expression analysis of long non-coding RNAs NKILA and PACER as well as their target genes, NF-κB and cox-2 in bipolar disorder patients. NUCLEOSIDES, NUCLEOTIDES & NUCLEIC ACIDS 2023; 42:527-537. [PMID: 36628999 DOI: 10.1080/15257770.2023.2166063] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/12/2023]
Abstract
Bipolar disorder (BD) is a severe condition characterized by periods of mania and depression. Despite advances in the neurobiology of bipolar disorder, the exact etiology of the disease remains unclear. There is evidence that Inflammation is associated with bipolar disorder. COX-2 and NF-κB are two critical mediators in the inflammatory pathways. Long non-coding RNAs (lncRNAs) are a new class of non-coding RNAs that play a wide range of roles, especially in developing and maintaining normal brain functions. Two lncRNAs called PACER and NKILA control the expression of COX-2 and NF-κB genes, respectively. In this study, Expression levels of PACER and NKILA lncRNAs, as well as, COX-2 and NF-κB genes were measured in fifty patients with bipolar disorder and 50 healthy individuals by real-time PCR. Expression levels of NKILA and COX2 were considerably reduced in BD patients compared with healthy controls. Such significant downregulation in the expression of NKILA and PACER was only observed in male patients with BD compared with male healthy subjects. Also, according to the results of the ROC curve, the area under curve values for NKILA and COX2 were 0.68 and 0.52 respectively. Consequently, the NKILA gene could be considered a biomarker. By examining the degree of pairwise correlation between genes, all genes had a significant positive correlation with each other. Taken together, these results revealed a function for NKILA and PACER lncRNAs in the pathogenesis of BD.
Collapse
Affiliation(s)
- Reza Mola-Ali-Nejad
- Department of Medical Genetics, School of Medicine, Jiroft University of Medical Sciences, Jiroft, Iran
| | - Saba Fakharianzadeh
- Department of Cell and Molecular Biology, Faculty of Life Sciences and Technology, Shahid Beheshti University, Tehran, Iran
| | - Zahra Maloum
- Department of Cell and Molecular Biology, Faculty of Life Sciences and Technology, Shahid Beheshti University, Tehran, Iran
| | - Mohammad Taheri
- Men's Health and Reproductive Health Research Center, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Zeinab Shirvani-Farsani
- Department of Cell and Molecular Biology, Faculty of Life Sciences and Technology, Shahid Beheshti University, Tehran, Iran
| |
Collapse
|
64
|
Chalcones: Promising therapeutic agents targeting key players and signaling pathways regulating the hallmarks of cancer. Chem Biol Interact 2023; 369:110297. [PMID: 36496109 DOI: 10.1016/j.cbi.2022.110297] [Citation(s) in RCA: 38] [Impact Index Per Article: 19.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2022] [Revised: 11/29/2022] [Accepted: 12/02/2022] [Indexed: 12/12/2022]
Abstract
The need for innovative anticancer treatments with high effectiveness and low toxicity is urgent due to the development of malignancies that are resistant to chemotherapeutic agents and the poor specificity of existing anticancer treatments. Chalcones are 1,3-diaryl-2-propen-1-ones, which are the precursors for flavonoids and isoflavonoids. Chalcones are readily available from a wide range of natural resources and consist of very basic chemical scaffolds. Because the ease with which the synthesis it allows for the production of several chalcone derivatives. Various in-vitro and in-vivo studies indicate that naturally occurring and synthetic chalcone derivatives exhibit promising biological activities against cancer hallmarks such as proliferation, angiogenesis, invasion, metastasis, inflammation, stemness, and regulation of cancer epigenetics. According to their structure and functional groups, chalcones derivatives and their hybrid compounds exert a broad range of biological activities through targeting key elements and signaling molecules relevant to cancer progression. This review will provide valuable insights into the latest updates of chalcone groups as anticancer agents and extensively discuss their underlying molecular mechanisms of action.
Collapse
|
65
|
Chan LP, Tseng YP, Wang HC, Chien CY, Wu CW, Wang LF, Liang CH. Growth Regulated Oncogene-α Upregulates TNF-α and COX-2 and Activates NOD1/RIPK2 mediated-MAPK Pathway in Head and Neck Squamous Cell Carcinoma. J Cancer 2023; 14:989-1000. [PMID: 37151389 PMCID: PMC10158519 DOI: 10.7150/jca.82300] [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: 01/01/2023] [Accepted: 03/25/2023] [Indexed: 05/09/2023] Open
Abstract
Purpose: The long-term prognosis and survival rate of patients with recurrent or metastatic head and neck squamous cell carcinoma (HNSCC) are poor, although the identification of specific biomarkers that reveal its nature and aggressiveness has improved it. Growth-related oncogene alpha (Groα) and NOD1 (nucleotide-binding oligomerization domain 1) can be used as prognosis markers to identify subgroups of HNSCC patients with low survival rates and as potential therapeutic targets for HNSCC patients. However, the mechanism associated with the Groα-mediated NOD pathway in HNSCC progression remains unclear. Method: Overall survival analysis and multiple-gene comparison were analyzed using Gene Expression Profiling Interactive Analysis (GEPIA). qRT-PCR and RT-PCR were used to analyze mRNA expression. Microarray, immunofluorescence staining or western blot analyses were carried out to detect protein expression. Results: Groα was significantly higher in the grade 4 HNSCC tumor tissues compared with that in grade 1-3 and healthy subjects. High expression of Groα, NOD1 and RIPK2 (receptor-interacting serine-threonine kinase 2) is correlated with survival rate in HNSCC patients. Treatment of SCC25 and OECM-1 cells with Groα increased the expression of NOD1 and RIPK2 in a concentration-dependent manner. The findings herein reveal the association of Groα, NOD1 and RIPK2 biomarkers with HNSCC carcinogenesis. Moreover, Groα is the major stimulus of inflammatory mediation and promotes TNF-α (tumor necrosis factor-α) and COX-2 (cyclooxygenase-2) expression in HNSCC. Groα induces TNF-α and COX-2 expression through regulation involving ERK (extracellular signal-regulated kinase)-, JNK (C-Jun N-terminal kinase)- and p38 MAPK (mitogen-activated protein kinase)-dependent signaling pathways. Conclusions: Our findings herein constitute the first evidence that Groα is important in HNSCC progression and metastasis via the NOD1-mediated MAPK pathway, suggesting a role for Groα and NOD1 in mediating metastasis and its potential as a therapeutic target.
Collapse
Affiliation(s)
- Leong-Perng Chan
- Department of Otorhinolaryngology-Head and Neck Surgery, Kaohsiung Medical University Hospital, Faculty of Medicine, College of Medicine, Kaohsiung Medical University, Kaohsiung, Taiwan
- Department of Otorhinolaryngology-Head and Neck Surgery, Kaohsiung Municipal Ta-Tung Hospital, Kaohsiung Medical University, Kaohsiung, Taiwan
- ✉ Corresponding authors: (L.-P. C.); (C.-H. L.); Tel.: +886-6-2664911 (ext. 2441)
| | - Ya-Ping Tseng
- Institute of Basic Medical Sciences, National Cheng Kung University, Tainan, Taiwan
| | - Hui-Ching Wang
- Department of Internal Medicine, Division of Hematology and Oncology, Kaohsiung Medical University Hospital, Kaohsiung Medical University, Kaohsiung, Taiwan
| | - Chen-Yu Chien
- Department of Otorhinolaryngology-Head and Neck Surgery, Kaohsiung Medical University Hospital, Faculty of Medicine, College of Medicine, Kaohsiung Medical University, Kaohsiung, Taiwan
- Department of Otolaryngology-Head and Neck Surgery, Kaohsiung Municipal Siaogang Hospital, Kaohsiung Medical University, Kaohsiung, Taiwan
| | - Che-Wei Wu
- Department of Otorhinolaryngology-Head and Neck Surgery, Kaohsiung Medical University Hospital, Faculty of Medicine, College of Medicine, Kaohsiung Medical University, Kaohsiung, Taiwan
| | - Ling-Feng Wang
- Department of Otorhinolaryngology-Head and Neck Surgery, Kaohsiung Medical University Hospital, Faculty of Medicine, College of Medicine, Kaohsiung Medical University, Kaohsiung, Taiwan
| | - Chia-Hua Liang
- Department of Cosmetic Science and Institute of Cosmetic Science, Chia Nan University of Pharmacy and Science, Tainan, Taiwan
- ✉ Corresponding authors: (L.-P. C.); (C.-H. L.); Tel.: +886-6-2664911 (ext. 2441)
| |
Collapse
|
66
|
Phytocannabinoids Act Synergistically with Non-Steroidal Anti-Inflammatory Drugs Reducing Inflammation in 2D and 3D In Vitro Models. Pharmaceuticals (Basel) 2022; 15:ph15121559. [PMID: 36559009 PMCID: PMC9787964 DOI: 10.3390/ph15121559] [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: 11/16/2022] [Revised: 12/05/2022] [Accepted: 12/12/2022] [Indexed: 12/23/2022] Open
Abstract
Lung inflammation is associated with elevated pro-inflammatory cytokines and chemokines. Treatment with FCBD:std (standard mix of cannabidiol [CBD], cannabigerol [CBG] and tetrahydrocannabivarin [THCV]) leads to a marked reduction in the inflammation of alveolar epithelial cells, but not in macrophages. In the present study, the combined anti-inflammatory effect of FCBD:std with two corticosteroids (dexamethasone and budesonide) and two non-steroidal anti-inflammatory drugs (NSAID; ibuprofen and diclofenac), was examined. Enzyme-linked immunosorbent assay (ELISA) was used to determine protein levels. Gene expression was determined by quantitative real-time PCR. Inhibition of cyclo-oxygenase (COX) activity was determined in vitro. FCBD:std and diclofenac act synergistically, reducing IL-8 levels in macrophages and lung epithelial cells. FCBD:std plus diclofenac also reduced IL-6, IL-8 and CCL2 expression levels in co-cultures of macrophages and lung epithelial cells, in 2D and 3D models. Treatment by FCBD:std and/or NSAID reduced COX-1 and COX-2 gene expression but not their enzymatic activity. FCBD:std and diclofenac exhibit synergistic anti-inflammatory effects on macrophages and lung epithelial cells, yet this combined activity needs to be examined in pre-clinical studies and clinical trials.
Collapse
|
67
|
Ephraim R, Feehan J, Fraser S, Nurgali K, Apostolopoulos V. Cancer Immunotherapy: The Checkpoint between Chronic Colitis and Colorectal Cancer. Cancers (Basel) 2022; 14:cancers14246131. [PMID: 36551617 PMCID: PMC9776998 DOI: 10.3390/cancers14246131] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2022] [Accepted: 12/07/2022] [Indexed: 12/15/2022] Open
Abstract
Inflammatory Bowel Disease (IBD) is a group of diseases that cause intestinal inflammation and lesions because of an abnormal immune response to host gut microflora. Corticosteroids, anti-inflammatories, and antibiotics are often used to reduce non-specific inflammation and relapse rates; however, such treatments are ineffective over time. Patients with chronic colitis are more susceptible to developing colorectal cancer, especially those with a longer duration of colitis. There is often a limit in using chemotherapy due to side effects, leading to reduced efficacy, leaving an urgent need to improve treatments and identify new therapeutic targets. Cancer immunotherapy has made significant advances in recent years and is mainly categorized as cancer vaccines, adoptive cellular immunotherapy, or immune checkpoint blockade therapies. Checkpoint markers are expressed on cancer cells to evade the immune system, and as a result checkpoint inhibitors have transformed cancer treatment in the last 5-10 years. Immune checkpoint inhibitors have produced long-lasting clinical responses in both single and combination therapies. Winnie mice are a viable model of spontaneous chronic colitis with immune responses like human IBD. Determining the expression levels of checkpoint markers in tissues from these mice will provide insights into disease initiation, progression, and cancer. Such information will lead to identification of novel checkpoint markers and the development of treatments with or without immune checkpoint inhibitors or vaccines to slow or stop disease progression.
Collapse
Affiliation(s)
- Ramya Ephraim
- Institute for Health and Sport, Victoria University, Melbourne, VIC 3030, Australia
| | - Jack Feehan
- Institute for Health and Sport, Victoria University, Melbourne, VIC 3030, Australia
- Australian Institute for Musculoskeletal Science, Melbourne, VIC 3021, Australia
| | - Sarah Fraser
- Institute for Health and Sport, Victoria University, Melbourne, VIC 3030, Australia
| | - Kulmira Nurgali
- Institute for Health and Sport, Victoria University, Melbourne, VIC 3030, Australia
- Australian Institute for Musculoskeletal Science, Melbourne, VIC 3021, Australia
| | - Vasso Apostolopoulos
- Institute for Health and Sport, Victoria University, Melbourne, VIC 3030, Australia
- Australian Institute for Musculoskeletal Science, Melbourne, VIC 3021, Australia
- Correspondence:
| |
Collapse
|
68
|
A transcriptome-based analysis reveals functional differences among Dendrobium officinale Kimura & Migo species from different growing regions and with different quality levels. MEDICINE IN NOVEL TECHNOLOGY AND DEVICES 2022. [DOI: 10.1016/j.medntd.2022.100163] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/19/2022] Open
|
69
|
Shaker AM, Shahin MI, AboulMagd AM, Abdel Aleem SA, Abdel-Rahman HM, Abou El Ella DA. Novel 1,3-diaryl pyrazole derivatives bearing methylsulfonyl moiety: Design, synthesis, molecular docking and dynamics, with dual activities as anti-inflammatory and anticancer agents through selectively targeting COX-2. Bioorg Chem 2022; 129:106143. [DOI: 10.1016/j.bioorg.2022.106143] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2022] [Revised: 08/28/2022] [Accepted: 09/06/2022] [Indexed: 12/20/2022]
|
70
|
Li L, Lu H, Zhang Y, Li Q, Shi S, Liu Y. Effect of Azelaic Acid on Psoriasis Progression Investigated Based on Phosphatidylinositol 3-Kinase (PI3K)/Protein Kinase B (AKT) Signaling Pathway. Clin Cosmet Investig Dermatol 2022; 15:2523-2534. [PMID: 36447569 PMCID: PMC9701457 DOI: 10.2147/ccid.s389760] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2022] [Accepted: 11/10/2022] [Indexed: 12/10/2023]
Abstract
OBJECTIVE To probe into the effect of azelaic acid on psoriasis based on the phosphatidylinositol 3-kinase (PI3K)/protein kinase B (AKT) signaling pathway. METHODS Psoriasis gene expression data were downloaded from the GEO database for differential expression analysis to identify differentially expressed genes (DEGs). KEGG and GSEA analyses were performed to identify important signaling pathways that may be involved in psoriasis progression for subsequent validation. Thirty-six C57BL/6 mice aged 8 weeks old were randomly assigned into the blank control group (n = 9), negative control group (n = 9), psoriasis model group (n = 9), and azelaic acid treat group (n = 9). Mice models of psoriasis were prepared with imiquimod (IMQ) in the latter two groups, and azelaic acid ointment was applied in azelaic acid treat group. Then, hematoxylin-eosin (HE) staining was carried out to detect the effect of azelaic acid on the pathological damage of mice models of psoriasis in each group. HaCaT cells cultured in vitro were divided into blank control group, negative control group (addition of azelaic acid), IL-17 group (20 ng/mL) and IL-17+azelaic acid group, with 3 replicates for each group. Immunofluorescence assay and Western blotting were used to detect the protein expression of PI3K/AKT signaling pathway related molecules. RESULTS KEGG analysis showed that DEGs were significantly enriched in PI3K-AKT signaling pathway. GSEA analysis showed that PI3K and MTOR signaling pathways were up-regulated in psoriasis, while AUTOPHAGY signaling pathway was down-regulated. HE staining showed that azelaic acid could significantly inhibit the local skin injury in mice caused by IMQ-induced psoriasis. Moreover, azelaic acid can inhibit the expression of PI3K/AKT signaling pathway related proteins phosphorylated (p)-PI3K, p-AKT, p-mammalian target of rapamycin (mTOR), vascular endothelial growth factor (VEGF), cyclooxygenase-2 (COX-2), angiogenin-1 and hypoxia-inducible factor-1α (HIF-1α). These results imply that azelaic acid may inhibit the activation of PI3K/AKT signaling pathway and angiogenesis, thereby improving the symptoms of psoriasis. CONCLUSION Azelaic acid may inhibit the activation of PI3K/AKT signaling pathway and angiogenesis, thereby improving the symptoms of psoriasis.
Collapse
Affiliation(s)
- Licui Li
- Department of Dermatology and Venereology, Hebei Medical University, Shijiazhuang, People’s Republic of China
- Department of Dermatology, Shijiazhuang Gaocheng People’s Hospital, Shijiazhuang, People’s Republic of China
| | - Huixiu Lu
- Department of Dermatology and Venereology, Hebei Medical University, Shijiazhuang, People’s Republic of China
- Department of Dermatology, Shijiazhuang People’s Hospital, Shijiazhuang, People’s Republic of China
| | - Yanli Zhang
- Department of Dermatology, The Third Hospital of Hebei Medical University, Shijiazhuang, People’s Republic of China
| | - Qian Li
- Department of Dermatology, The Third Hospital of Hebei Medical University, Shijiazhuang, People’s Republic of China
| | - Shaomin Shi
- Department of Dermatology, The Third Hospital of Hebei Medical University, Shijiazhuang, People’s Republic of China
| | - Yaling Liu
- Department of Dermatology and Venereology, Hebei Medical University, Shijiazhuang, People’s Republic of China
- Department of Dermatology, The Third Hospital of Hebei Medical University, Shijiazhuang, People’s Republic of China
| |
Collapse
|
71
|
Is Caperatic Acid the Only Compound Responsible for Activity of Lichen Platismatia glauca within the Nervous System? Antioxidants (Basel) 2022; 11:antiox11102069. [PMID: 36290793 PMCID: PMC9598164 DOI: 10.3390/antiox11102069] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2022] [Revised: 10/13/2022] [Accepted: 10/14/2022] [Indexed: 11/26/2022] Open
Abstract
Lichens are a source of various biologically active compounds. However, the knowledge about them is still scarce, and their use in medicine is limited. This study aimed to investigate the therapeutic potential of the lichen Platismatia glauca and its major metabolite caperatic acid in regard to their potential application in the treatment of central nervous system diseases, especially neurodegenerative diseases and brain tumours, such as glioblastoma. First, we performed the phytochemical analysis of the tested P. glauca extracts based on FT-IR derivative spectroscopic and gas chromatographic results. Next the antioxidant properties were determined, and moderate anti-radical activity, strong chelating properties of Cu2+ and Fe2+ ions, and a mild effect on the antioxidant enzymes of the tested extracts and caperatic acid were proved. Subsequently, the influence of the tested extracts and caperatic acid on cholinergic transmission was determined by in vitro and in silico studies confirming that inhibitory effect on butyrylcholinesterase is stronger than against acetylcholinesterase. We also confirmed the anti-inflammatory properties of P. glauca extracts and caperatic acid using a COX-2 and hyaluronidase inhibition models. Moreover, our studies show the cytotoxic and pro-apoptotic activity of the P. glauca extracts against T98G and U-138 MG glioblastoma multiforme cell lines. In conclusion, it is possible to assume that P. glauca extracts and especially caperatic acid can be regarded as the source of the valuable substances to finding new therapies of central nervous system diseases.
Collapse
|
72
|
Design Strategies and Precautions for Using Vaccinia Virus in Tumor Virotherapy. Vaccines (Basel) 2022; 10:vaccines10091552. [PMID: 36146629 PMCID: PMC9504998 DOI: 10.3390/vaccines10091552] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2022] [Revised: 08/27/2022] [Accepted: 09/10/2022] [Indexed: 11/17/2022] Open
Abstract
Oncolytic virotherapy has emerged as a novel form of cancer immunotherapy. Oncolytic viruses (OVs) can directly infect and lyse the tumor cells, and modulate the beneficial immune microenvironment. Vaccinia virus (VACV) is a promising oncolytic vector because of its high safety, easy gene editing, and tumor intrinsic selectivity. To further improve the safety, tumor-targeting ability, and OV-induced cancer-specific immune activation, various approaches have been used to modify OVs. The recombinant oncolytic VACVs with deleting viral virulence factors and/or arming various therapeutic genes have displayed better therapeutic effects in multiple tumor models. Moreover, the combination of OVs with other cancer immunotherapeutic approaches, such as immune checkpoint inhibitors and CAR-T cells, has the potential to improve the outcome in cancer patients. This will open up new possibilities for the application of OVs in cancer treatment, especially for personalized cancer therapies.
Collapse
|
73
|
Inflammation- and Metastasis-Related Proteins Expression Changes in Early Stages in Tumor and Non-Tumor Adjacent Tissues of Colorectal Cancer Samples. Cancers (Basel) 2022; 14:cancers14184487. [PMID: 36139645 PMCID: PMC9497293 DOI: 10.3390/cancers14184487] [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: 08/04/2022] [Revised: 09/05/2022] [Accepted: 09/14/2022] [Indexed: 11/17/2022] Open
Abstract
Simple Summary Non-tumor adjacent tissue plays a key role in colorectal cancer development, as well as chronic inflammation, but their role has not yet been dilucidated. In addition, inflammation is a process which is related to epithelial-mesenchymal transition and metastasis, but their changes across the different colorectal cancer stages are not fully studied. Understanding how these processes participate in all colorectal cancer phases can be key to a better understanding of the disease. Abstract Chronic inflammation can induce malignant cell transformation, having an important role in all colorectal cancer (CRC) phases. Non-tumor adjacent tissue plays an important role in tumor progression, but its implication in CRC has not yet been fully elucidated. The aim was to analyze the expression of inflammatory, epithelial-mesenchymal transition (EMT), and metastasis-related proteins in both tumor and non-tumor adjacent tissues from CRC patients by western blot. Tumor tissue presented an increase in metastasis and EMT-related proteins compared to non-tumor adjacent tissue, especially in stage II. Tumor tissue stage II also presented an increase in inflammatory-related proteins compared to other stages, which was also seen in non-tumor adjacent tissue stage II. Additionally, the relapse-free survival study of Vimentin and VEGF-B expression levels in stage II patients showed that the higher the expression levels of each protein, the lower 10-year relapse-free survival. These could suggest that some metastasis-related signalling pathways may be activated in stage II in tumor tissue, accompanied by an increase in inflammation. Furthermore, non-tumor adjacent tissue presented an increase of the inflammatory status that could be the basis for future tumor progression. In conclusion, these proteins could be useful as biomarkers of diagnosis for CRC at early stages.
Collapse
|
74
|
Lin CT, Lin CF, Wu JT, Tsai HP, Cheng SY, Liao HJ, Lin TC, Wu CH, Lin YC, Wang JH, Chang GR. Effects of Para-Toluenesulfonamide on Canine Melanoma Xenotransplants in a BALB/c Nude Mouse Model. Animals (Basel) 2022; 12:2272. [PMID: 36077992 PMCID: PMC9454485 DOI: 10.3390/ani12172272] [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: 06/15/2022] [Revised: 08/05/2022] [Accepted: 08/09/2022] [Indexed: 11/17/2022] Open
Abstract
The pharmacological pathway of para-toluenesulfonamide (PTS) restricts the kinase activity of the mammalian target of rapamycin, potentially leading to reductions in cell division, cell growth, cell proliferation, and inflammation. These pathways have a critical effect on tumorigenesis. We aimed to examine the antitumor effect of PTS or PTS combined with cisplatin on canine melanoma implanted in BALB/c nude mice by estimating tumor growth, apoptosis expression, inflammation, and metastasis. The mice were randomly divided into four groups: control, cisplatin, PTS, and PTS combined with cisplatin. Mice treated with PTS or PTS combined with cisplatin had retarded tumor growth and increased tumor apoptosis through the enhanced expression of cleaved caspase 3 and extracellular signal-regulated kinase phosphorylation, decreased inflammatory cytokine levels, reduced inflammation-related factors, enhanced anti-inflammation-related factors, and inhibition of metastasis-related factors. Mice treated with PTS combined with cisplatin exhibited significantly retarded tumor growth, reduced tumor size, and increased tumor inhibition compared with those treated with cisplatin or PTS alone. PTS or PTS combined with cisplatin could retard canine melanoma growth and inhibit tumorigenesis. PTS and cisplatin were found to have an obvious synergistic tumor-inhibiting effect on canine melanoma. PTS alone and PTS combined with cisplatin may be antitumor agents for canine melanoma treatment.
Collapse
Affiliation(s)
- Chien-Teng Lin
- Ph.D. Program of Agriculture Science, National Chiayi University, 300 University Road, Chiayi 60004, Taiwan
- Department of Veterinary Medicine, National Chiayi University, 580 Xinmin Road, Chiayi 60054, Taiwan
| | - Chuen-Fu Lin
- Department of Veterinary Medicine, College of Veterinary Medicine, National Pingtung University of Science and Technology, 1 Shuefu Road, Neipu, Pingtung 912301, Taiwan
| | - Jui-Te Wu
- Department of Veterinary Medicine, National Chiayi University, 580 Xinmin Road, Chiayi 60054, Taiwan
| | - Hsiao-Pei Tsai
- Ph.D. Program of Agriculture Science, National Chiayi University, 300 University Road, Chiayi 60004, Taiwan
- Department of Veterinary Medicine, National Chiayi University, 580 Xinmin Road, Chiayi 60054, Taiwan
| | - Shu-Ying Cheng
- Department of Veterinary Medicine, National Chiayi University, 580 Xinmin Road, Chiayi 60054, Taiwan
- Department of Pet Medicine, Gongwin Biopharma Co., Ltd., 1 Section, 80 Jianguo North Road, Zhongshan District, Taipei 104001, Taiwan
| | - Huei-Jyuan Liao
- Department of Veterinary Medicine, National Chiayi University, 580 Xinmin Road, Chiayi 60054, Taiwan
| | - Tzu-Chun Lin
- Department of Veterinary Medicine, National Chiayi University, 580 Xinmin Road, Chiayi 60054, Taiwan
| | - Chao-Hsuan Wu
- Department of Veterinary Medicine, National Chiayi University, 580 Xinmin Road, Chiayi 60054, Taiwan
- Department of Pet Medicine, Gongwin Biopharma Co., Ltd., 1 Section, 80 Jianguo North Road, Zhongshan District, Taipei 104001, Taiwan
| | - Yu-Chin Lin
- Department of Veterinary Medicine, National Chiayi University, 580 Xinmin Road, Chiayi 60054, Taiwan
- Department of Pet Medicine, Gongwin Biopharma Co., Ltd., 1 Section, 80 Jianguo North Road, Zhongshan District, Taipei 104001, Taiwan
| | - Jiann-Hsiung Wang
- Department of Veterinary Medicine, National Chiayi University, 580 Xinmin Road, Chiayi 60054, Taiwan
| | - Geng-Ruei Chang
- Department of Veterinary Medicine, National Chiayi University, 580 Xinmin Road, Chiayi 60054, Taiwan
| |
Collapse
|
75
|
Song X, Greiner-Tollersrud OK, Zhou H. Oral Microbiota Variation: A Risk Factor for Development and Poor Prognosis of Esophageal Cancer. Dig Dis Sci 2022; 67:3543-3556. [PMID: 34505256 DOI: 10.1007/s10620-021-07245-2] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/28/2021] [Accepted: 08/23/2021] [Indexed: 12/24/2022]
Abstract
Recent studies have shown that oral microbiota play an important role in the esophageal cancer (EC) initiation and progression, suggesting that oral microbiota is a new risk factor for EC. The composition of the microbes inhabiting the oral cavity could be perturbed with continuous factors such as smoking, alcohol consumption, and inflammation. The microbial alteration involves the decrease of beneficial species and the increase of pathogenic species. Experimental evidences suggest a significant role of oral commensal organisms in protecting hosts against EC. By contrast, oral pathogens, especially Porphyromonas gingivalis and Fusobacterium nucleatum, give rise to the risk for developing EC through their pro-inflammatory and pro-tumorigenic activities. The presences of oral dysbiosis, microbial biofilm, and periodontitis in EC patients are found to be associated with invasive cancer phenotypes and poor prognosis. The mechanism of oral bacteria in EC progression is complex, which involves a combination of cytokines, chemokines, oncogenic signaling pathways, cell surface receptors, the degradation of extracellular matrix, and cell apoptosis. From a clinical perspective, good oral hygiene, professional oral care, and rational use of antibiotics bring positive impacts on oral microbial balance, thus helping individuals reduce the risk of EC, inhibiting postoperative complications among EC patients, and improving the efficiency of chemoradiotherapy. However, current oral hygiene practices mainly focus on the oral bacteria-based predictive and preventive purposes. It is still far from implementing microbiota-dependent regulation as a therapy for EC. Further explorations are needed to render oral microbiota a potential target for treating EC.
Collapse
Affiliation(s)
- Xiaobo Song
- Department of Microbiology, Dalian Medical University, No.9 West Section Lvshun South Road, Dalian, 116044, Liaoning Province, China.,Department of Medical Biology, Faculty of Health Sciences, University of Tromsø, 9037, Tromsø, Norway
| | - Ole K Greiner-Tollersrud
- Department of Medical Biology, Faculty of Health Sciences, University of Tromsø, 9037, Tromsø, Norway
| | - Huimin Zhou
- Department of Microbiology, Dalian Medical University, No.9 West Section Lvshun South Road, Dalian, 116044, Liaoning Province, China.
| |
Collapse
|
76
|
Li J, Hu X, Zhang H, Peng Y, Li S, Xiong Y, Jiang W, Wang Z. N-2-(Phenylamino) Benzamide Derivatives as Dual Inhibitors of COX-2 and Topo I Deter Gastrointestinal Cancers via Targeting Inflammation and Tumor Progression. J Med Chem 2022; 65:10481-10505. [PMID: 35868003 DOI: 10.1021/acs.jmedchem.2c00635] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
Abstract
Given the close association between inflammation and cancer, combining anti-inflammation therapy is prominent to improve the anticancer effect. Based on I-1, a series of agents targeting COX-2 and Topo I were designed by combining fenamates and phenols. The optimal compound 1H-30 displayed an enhanced inhibitory effect on COX-2 compared to tolfenamic acid and I-1 and showed better inhibition of Topo I than I-1. Importantly, 1H-30 showed potential anticancer effects and suppressed the activation of the NF-κB pathway in cancer cells. 1H-30 inhibited the nuclear translocation of NF-κB and suppressed the production of NO, COX-2, and IL-1β in RAW264.7. In vivo, 1H-30 showed acceptable pharmacokinetic parameters, decreased the tumor growth without affecting the body weight, down-regulated COX-2 and MMP-9, and induced apoptosis in the CT26.WT tumor-bearing mice. Accordingly, 1H-30 as a potential Topo I/COX-2 inhibitor which possessed anti-inflammatory and anticancer effects, with inhibition of the NF-κB pathway, is promising for gastrointestinal cancer therapy.
Collapse
Affiliation(s)
- Junfang Li
- School of Pharmacy, Lanzhou University, Lanzhou 730000, China.,State Key Laboratory of Applied Organic Chemistry, College of Chemistry and Chemical Engineering, Lanzhou University, Lanzhou 730000, China
| | - Xiaoling Hu
- School of Pharmacy, Lanzhou University, Lanzhou 730000, China.,State Key Laboratory of Applied Organic Chemistry, College of Chemistry and Chemical Engineering, Lanzhou University, Lanzhou 730000, China
| | - Honghua Zhang
- School of Pharmacy, Lanzhou University, Lanzhou 730000, China.,State Key Laboratory of Applied Organic Chemistry, College of Chemistry and Chemical Engineering, Lanzhou University, Lanzhou 730000, China
| | - Yan Peng
- School of Pharmaceutical Science, Hengyang Medical School, University of South China, Hengyang 421001, Hunan, China
| | - Shuang Li
- School of Pharmaceutical Science, Hengyang Medical School, University of South China, Hengyang 421001, Hunan, China
| | - Yongxia Xiong
- School of Pharmaceutical Science, Hengyang Medical School, University of South China, Hengyang 421001, Hunan, China
| | - Weifan Jiang
- School of Pharmaceutical Science, Hengyang Medical School, University of South China, Hengyang 421001, Hunan, China
| | - Zhen Wang
- School of Pharmaceutical Science, Hengyang Medical School, University of South China, Hengyang 421001, Hunan, China.,School of Pharmacy, Lanzhou University, Lanzhou 730000, China.,State Key Laboratory of Applied Organic Chemistry, College of Chemistry and Chemical Engineering, Lanzhou University, Lanzhou 730000, China
| |
Collapse
|
77
|
Kumar P, Sharma R, Garg N. Withania somnifera - a magic plant targeting multiple pathways in cancer related inflammation. PHYTOMEDICINE : INTERNATIONAL JOURNAL OF PHYTOTHERAPY AND PHYTOPHARMACOLOGY 2022; 101:154137. [PMID: 35533610 DOI: 10.1016/j.phymed.2022.154137] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/16/2021] [Revised: 03/31/2022] [Accepted: 04/28/2022] [Indexed: 06/14/2023]
Abstract
BACKGROUND Deregulated inflammatory responses are known to play a pivotal role in cancer initiation and progression. Tumor microenvironment is associated with the presence of a diverse array of inflammatory reactions, which further help tumor growth, metastasis and drug resistance. Withania somnifera is known to curb proliferation of cancer cells and lower inflammatory responses. PURPOSE In order to minimize the inflammation, cancer treatments often include immunomodulatory drugs. However, given the side effects of both of the cytotoxic cancer drugs and synthetic immunomodulatory agents, there is a need to develop novel anti-inflammatory agents for improved cancer therapy. A number of reports indicate that bioactive phytochemicals derived from W. somnifera exhibit anti-inflammatory capabilities in cancer. A deeper look into the underlying molecular mechanisms implicated in W. somnifera mediated anti inflammation is lacking, which is essential to fully understand the potential of this magical plant in cancer. Therefore, in the present review we are summarizing various reports, which describe mechanistic understanding of W. somnifera in cancer related inflammation. STUDY DESIGN AND METHODOLOGY In order to gather information on the molecular pathways affected by W. somnifera in cancer related inflammation, 'PubMed' and 'Science Direct' databases were searched using keywords Withania, cancer inflammation, and Withaferin A. Selected literature was analyzed to cover the role of inflammation in cancer, usage and side effects of anti-inflammatory drugs, W. somnifera as an immunomodulatory agent in cancer, molecular pathways modulated by W. somnifera in various preclinical models, and clinical trials using W. somnifera as an anti-inflammatory agent. RESULTS Upon literature survey we found that both W. somnifera extracts and Withaferin-A, exhibit anti inflammatory activities in various preclinical cancer models. W. somnifera modulates a number of signaling pathways such as NF-kB, JAK-STAT and AP1 to reduce cancer related inflammation. Anti inflammatory properties of W. somnifera might be effective in the treatment of drug resistance in cancers. Based on its promising effects against cancer associated inflammation in preclinical studies, W. somnifera derived products are being tested in clinical trials. CONCLUSION Several preclinical studies demonstrated anti-inflammatory potential of W. somnifera in a variety of cancers. While a few clinical trials are investigating the role of W. somnifera in various diseases, focused studies on its role in cancer related inflammation are lacking. Additionally, its anti-inflammatory effects offer targeting of senescence associated secretory phenotype (SASP), which is speculated to play a critical role in chemoresistance. Apart from targeting cancer cell proliferation, anti-inflammatory effects of Withania provide double advantage in cancer management. Therefore, clinical trials to target cancer related inflammation using W. somnifera as a drug, should be performed to validate its advantages in cancer therapy.
Collapse
Affiliation(s)
- Praveen Kumar
- Department of Medicinal Chemistry, Faculty of Ayurveda, Institute of Medical Sciences, Banaras Hindu University, Varanasi, Uttar Pradesh 221005, India; Department of Rasa Shastra & Bhaishajya Kalpana, Faculty of Ayurveda, Institute of Medical Sciences, Banaras Hindu University, Varanasi, Uttar Pradesh 221005, India
| | - Rohit Sharma
- Department of Rasa Shastra & Bhaishajya Kalpana, Faculty of Ayurveda, Institute of Medical Sciences, Banaras Hindu University, Varanasi, Uttar Pradesh 221005, India
| | - Neha Garg
- Department of Medicinal Chemistry, Faculty of Ayurveda, Institute of Medical Sciences, Banaras Hindu University, Varanasi, Uttar Pradesh 221005, India.
| |
Collapse
|
78
|
Nascente EDP, Amorim RL, Fonseca-Alves CE, de Moura VMBD. Comparative Pathobiology of Canine and Human Prostate Cancer: State of the Art and Future Directions. Cancers (Basel) 2022; 14:2727. [PMID: 35681707 PMCID: PMC9179314 DOI: 10.3390/cancers14112727] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2022] [Revised: 05/14/2022] [Accepted: 05/15/2022] [Indexed: 02/01/2023] Open
Abstract
First described in 1817, prostate cancer is considered a complex neoplastic entity, and one of the main causes of death in men in the western world. In dogs, prostatic carcinoma (PC) exhibits undifferentiated morphology with different phenotypes, is hormonally independent of aggressive character, and has high rates of metastasis to different organs. Although in humans, the risk factors for tumor development are known, in dogs, this scenario is still unclear, especially regarding castration. Therefore, with the advent of molecular biology, studies were and are carried out with the aim of identifying the main molecular mechanisms and signaling pathways involved in the carcinogenesis and progression of canine PC, aiming to identify potential biomarkers for diagnosis, prognosis, and targeted treatment. However, there are extensive gaps to be filled, especially when considering the dog as experimental model for the study of this neoplasm in humans. Thus, due to the complexity of the subject, the objective of this review is to present the main pathobiological aspects of canine PC from a comparative point of view to the same neoplasm in the human species, addressing the historical context and current understanding in the scientific field.
Collapse
Affiliation(s)
- Eduardo de Paula Nascente
- School of Veterinary Medicine and Animal Science, Federal University of Goiás, Goiânia 74001-970, Brazil;
| | - Renée Laufer Amorim
- Veterinary Clinic Department, School of Veterinary Medicine and Animal Science, São Paulo State University (UNESP), Botucatu 18618-970, Brazil;
| | - Carlos Eduardo Fonseca-Alves
- Department of Veterinary Surgery and Anesthesiology, School of Veterinary Medicine and Animal Science, São Paulo State University (UNESP), Botucatu 18618-970, Brazil;
| | | |
Collapse
|
79
|
A Novel Herbal Extract Blend Product Prevents Particulate Matters-Induced Inflammation by Improving Gut Microbiota and Maintaining the Integrity of the Intestinal Barrier. Nutrients 2022; 14:nu14102010. [PMID: 35631153 PMCID: PMC9145798 DOI: 10.3390/nu14102010] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2022] [Revised: 04/26/2022] [Accepted: 05/07/2022] [Indexed: 02/05/2023] Open
Abstract
Air pollutants of PM2.5 can alter the composition of gut microbiota and lead to inflammation in the lung and gastrointestinal tract. The aim of this study was to evaluate the protective effect of a novel herbal extract blend, FC, composed of Lonicera japonica extract, Momordica grosvenori extract, and broccoli seed extract, on PM2.5-induced inflammation in the respiratory and intestinal tract. A549 cells and THP-1 cells, as well as C57BL/6 mice, were stimulated with PM2.5 to establish in vitro and in vivo exposure models. The models were treated with or without FC. The expression of inflammatory cytokines and tight junction proteins were studied. Proteomic analysis was performed to elucidate mechanisms. Mouse feces were collected for gut microbiota analysis. FC was shown to modulate the upregulation of pro-inflammatory cytokines mRNA expression in A549 and THP-1 cells and downregulated tight junction proteins mRNA expression in A549 cells due to PM2.5 stimulation. In animal models, the decreased expression of the anti-inflammatory factor il-10, tight junction protein ZO-1, and the elevated expression of COX-2 induced by PM2.5 were improved by FC intervention, which may be associated with zo-1 and cox-2 signaling pathways. In addition, FC was shown to improve the gut microbiota by increasing the abundance of beneficial bacteria.
Collapse
|
80
|
Ansari MA, Thiruvengadam M, Venkidasamy B, Alomary MN, Salawi A, Chung IM, Shariati MA, Rebezov M. Exosome-based nanomedicine for cancer treatment by targeting inflammatory pathways: Current status and future perspectives. Semin Cancer Biol 2022; 86:678-696. [PMID: 35452820 DOI: 10.1016/j.semcancer.2022.04.005] [Citation(s) in RCA: 35] [Impact Index Per Article: 11.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2021] [Revised: 02/23/2022] [Accepted: 04/14/2022] [Indexed: 12/12/2022]
Abstract
Cancer is one of the dreadful diseases worldwide. Surgery, radiation and chemotherapy, are the three basic standard modes of cancer treatment. However, difficulties in cancer treatment are increasing due to immune escape, spreading of cancer to other places, and resistance of cancer cells to therapies. Various signaling mechanisms, including PI3K/Akt/mTOR, RAS, WNT/β-catenin, TGF-beta, and notch pathways, are involved in cancer resistance. The adaptive inflammatory response is the initial line of defence against infection. However, chronic inflammation can lead to tumorigenesis, malignant transformation, tumor growth, invasion, and metastasis. The most commonly dysregulated inflammatory pathways linked to cancer include NF-κB, MAPK, JAK-STAT, and PI3K/AKT. To overcome major hurdles in cancer therapy, nanomedicine is receiving much attention due to its role as a vehicle for delivering chemotherapeutic agents that specifically target tumor sites. Several biocompatible nanocarriers including polymer and inorganic nanoparticles, liposomes, micellar nanoparticles, nanotubes, and exosomes have been extensively studied. Exosome has been reported as an important potential sytem that could be effectively used as a bioinspired, bioengineered, and biomimetic drug delivery solution considering its toxicity, immunogenicity, and rapid clearance by the mononuclear phagocyte system. Exosome-mimetic vesicles are receiving much interest for developing nano-sized delivery systems. In this review, exosomes in detail as well as certain other nanocarriers, and their potential therapeutic roles in cancer therapy has been thoroughly discussed. Additionally, we also reviewed on oncogenic and tumor suppressor proteins, inflammation, and their associated signaling pathways and their interference by exosomes based nanomedicine.
Collapse
Affiliation(s)
- Mohammad Azam Ansari
- Department of Epidemic Disease Research, Institutes for Research and Medical Consultations (IRMC), Imam Abdulrahman Bin Faisal University, Dammam 31441, Saudi Arabia
| | - Muthu Thiruvengadam
- Department of Crop Science, College of Sanghuh Life Science, Konkuk University, Seoul 05029, Republic of Korea.
| | - Baskar Venkidasamy
- Department of Biotechnology, Sri Shakthi Institute of Engineering and Technology, Coimbatore 641062, Tamil Nadu, India
| | - Mohammad N Alomary
- National Centre for Biotechnology, King Abdulaziz City for Science and Technology (KACST), P.O. Box 6086, Riyadh 11442, Saudi Arabia
| | - Ahmad Salawi
- Department of Pharmaceutics, College of Pharmacy, Jazan University, Jazan, Saudi Arabia
| | - Ill-Min Chung
- Department of Crop Science, College of Sanghuh Life Science, Konkuk University, Seoul 05029, Republic of Korea.
| | - Mohammad Ali Shariati
- Research Department, K.G. Razumovsky Moscow State University of Technologies and Management (The First Cossack University), 73, Zemlyanoy Val St., Moscow 109004, Russian Federation
| | - Maksim Rebezov
- Department of Scientific Advisers, V. M. Gorbatov Federal Research Center for Food Systems, 26 Talalikhina St., Moscow 109316, Russian Federation
| |
Collapse
|
81
|
Qin J, Song J, Liang Y, Jiao A, Yang B. In-silico analysis identifies the anti-liver injury targets of diammonium glycyrrhizinate: validated in perfluorooctanoic acid-lesioned mouse model. Chem Biodivers 2022; 19:e202100938. [PMID: 35362201 DOI: 10.1002/cbdv.202100938] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2021] [Accepted: 03/31/2022] [Indexed: 11/05/2022]
Abstract
Liver injury refers to a pathological condition that causes dysfunction to hepatic parenchymal cells. And diammonium glycyrrhizinate (DG) is clinically prescribed for hepatoprotection. To date, detailed information regarding DG against liver injury in molecular mechanisms remains unrevealed totally. In the present study, we applied network pharmacology and molecular docking to decipher substantial genes, biological functions of DG for treating liver injury. Furthermore, preclinical experiments using perfluorooctanoic acid (PFOA)-induced liver injury in mice were used to validate the bioinformatic findings. Our results showed that the target network of DG and liver injury predominantly shared 90 genes. Eleven core genes of DG treating liver injury including ALB, TP53, TNF, CASP3, PTGS2, JUN, TLR4, IL10, STAT3, NOS3, FOS. The gene ontology and KEGG enrichment further highlighted their importance in regulation of cell proliferation, regulation of transcription, inflammatory response, regulation of NF-kappaB import into nucleus, regulation of apoptotic process, T cell receptor signaling pathway, and Toll-like receptor signaling pathway. Moreover, DG treatment was found to rescue the PFOA-induced liver injury through the modulation of identified genes including TNF, CASP3, PTGS2, and ALB. Current integrated data from bioinformatics method and experimental validation uncovered that DG exerts potent actions to treat liver injury through regulating core targets associated with inflammation and immunomodulation.
Collapse
Affiliation(s)
- Jingru Qin
- Guangxi Medical University, College of Pharmacy, Shuangyong Road No.22, Nanning, CHINA
| | - Jianhua Song
- guangxi medical university, TCM, Zhongshan Road No 1, Guigang, CHINA
| | - Yujia Liang
- Guangxi Medical University, TCM, Shuangyong Road No.22, Nanning, CHINA
| | - Aijun Jiao
- Guangxi Medical University, TCM, Shuangyong Road No.22, Nanning, CHINA
| | - Bin Yang
- Guangxi Medical University, College of Pharmacy, Shuangyong Road No.22, 541004, Nanning, CHINA
| |
Collapse
|
82
|
Zhang F, Qiao S. Research Progress on the Relationship Between Inflammation and Colorectal Cancer. Ann Gastroenterol Surg 2022; 6:204-211. [PMID: 35261946 PMCID: PMC8889855 DOI: 10.1002/ags3.12517] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/18/2021] [Revised: 09/15/2021] [Accepted: 09/29/2021] [Indexed: 02/06/2023] Open
Abstract
Colorectal cancer is one of the common malignant tumors. Relevant epidemiology and a large number of experimental studies have proved that chronic inflammation is highly correlated with the occurrence and development of colorectal cancer. And inflammatory bowel disease has been proven to be an independent risk factor for colorectal cancer. Various inflammatory cells participate in the establishment of the chronic inflammatory intestinal microenvironment required for the onset of colorectal cancer. The abnormal signal pathways mediated by various inflammatory factors and inflammatory mediators promote the occurrence of tumors, which are related to colorectal cancer and pathogenesis-related inflammation mechanisms. At the gene level, miRNAs can also affect the pathogenesis of colorectal cancer by regulating mesenchymal epithelial transformation. This article reviews the relationship between inflammation and colorectal cancer as well as the related inflammatory mechanisms.
Collapse
Affiliation(s)
- Feng Zhang
- Department of General SurgeryTongren Municipal People’s Hospital of Guizhou Medical University (GMU)GuizhouChina
| | - Song Qiao
- Department of General SurgeryTongren Municipal People’s Hospital of Guizhou Medical University (GMU)GuizhouChina
| |
Collapse
|
83
|
Hussain Y, Alam W, Ullah H, Dacrema M, Daglia M, Khan H, Arciola CR. Antimicrobial Potential of Curcumin: Therapeutic Potential and Challenges to Clinical Applications. Antibiotics (Basel) 2022; 11:322. [PMID: 35326785 PMCID: PMC8944843 DOI: 10.3390/antibiotics11030322] [Citation(s) in RCA: 78] [Impact Index Per Article: 26.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2022] [Revised: 02/25/2022] [Accepted: 02/25/2022] [Indexed: 02/06/2023] Open
Abstract
Curcumin is a bioactive compound that is extracted from Curcuma longa and that is known for its antimicrobial properties. Curcuminoids are the main constituents of curcumin that exhibit antioxidant properties. It has a broad spectrum of antibacterial actions against a wide range of bacteria, even those resistant to antibiotics. Curcumin has been shown to be effective against the microorganisms that are responsible for surgical infections and implant-related bone infections, primarily Staphylococcus aureus and Escherichia coli. The efficacy of curcumin against Helicobacter pylori and Mycobacterium tuberculosis, alone or in combination with other classic antibiotics, is one of its most promising antibacterial effects. Curcumin is known to have antifungal action against numerous fungi that are responsible for a variety of infections, including dermatophytosis. Candidemia and candidiasis caused by Candida species have also been reported to be treated using curcumin. Life-threatening diseases and infections caused by viruses can be counteracted by curcumin, recognizing its antiviral potential. In combination therapy with other phytochemicals, curcumin shows synergistic effects, and this approach appears to be suitable for the eradication of antibiotic-resistant microbes and promising for achieving co-loaded antimicrobial pro-regenerative coatings for orthopedic implant biomaterials. Poor water solubility, low bioavailability, and rapid degradation are the main disadvantages of curcumin. The use of nanotechnologies for the delivery of curcumin could increase the prospects for its clinical application, mainly in orthopedics and other surgical scenarios. Curcumin-loaded nanoparticles revealed antimicrobial properties against S. aureus in periprosthetic joint infections.
Collapse
Affiliation(s)
- Yaseen Hussain
- College of Pharmaceutical Sciences, Soochow University, Suzhou 215123, China;
- Department of Pharmacy, Abdul Wali Khan University Mardan, Mardan 23200, Pakistan;
| | - Waqas Alam
- Department of Pharmacy, Abdul Wali Khan University Mardan, Mardan 23200, Pakistan;
| | - Hammad Ullah
- Department of Pharmacy, University of Naples Federico II, 80131 Naples, Italy; (H.U.); (M.D.)
| | - Marco Dacrema
- Department of Pharmacy, University of Naples Federico II, 80131 Naples, Italy; (H.U.); (M.D.)
| | - Maria Daglia
- Department of Pharmacy, University of Naples Federico II, 80131 Naples, Italy; (H.U.); (M.D.)
- International Research Center for Food Nutrition and Safety, Jiangsu University, Zhenjiang 212013, China
| | - Haroon Khan
- Department of Pharmacy, Abdul Wali Khan University Mardan, Mardan 23200, Pakistan;
| | - Carla Renata Arciola
- Laboratorio di Patologia delle Infezioni Associate all’Impianto, IRCCS Istituto Ortopedico Rizzoli, Via di Barbiano 1/10, 40136 Bologna, Italy;
- Laboratorio di Immunoreumatologia e Rigenerazione Tissutale, IRCCS Istituto Ortopedico Rizzoli, Via di Barbiano 1/10, 40136 Bologna, Italy
- Department of Experimental, Diagnostic and Specialty Medicine, University of Bologna, Via San Giacomo 14, 40136 Bologna, Italy
| |
Collapse
|
84
|
Meloxicam Inhibits Hepatocellular Carcinoma Progression and Enhances the Sensitivity of Immunotherapy via the MicroRNA-200/PD-L1 Pathway. JOURNAL OF ONCOLOGY 2022; 2022:4598573. [PMID: 35237322 PMCID: PMC8885196 DOI: 10.1155/2022/4598573] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 11/27/2021] [Revised: 01/12/2022] [Accepted: 01/28/2022] [Indexed: 12/14/2022]
Abstract
Background. Hepatocellular carcinoma (HCC) has become the sixth most common cancer and the third leading cause of cancer death in the world. Although the research achievements of tumor immunotherapy have made great progress, especially the combination of immune targeted therapy has achieved good curative effect in HCC, but only a few patients are suitable for it and benefit from it. Therefore, there is an urgent need to find new effective drugs to treat HCC or to enhance the sensitivity of immunotherapy. Methods. Meloxicam, a COX2 inhibitor with strong anti-HCC potential, was screened from 800 small molecules approved by FDA. The effect of meloxicam on the proliferation, invasion, and migration of HCC cell lines was evaluated by cell phenotype analysis. The Human Protein Atlas database and the TISCH database were used to analyze COX2 data in single cells, and the TISIDB database was used to analyze the correlation of COX2 with immune function. The real-time quantitative polymerase chain reaction (qRT-PCR) and western blot were used to evaluate the level of PD-L1 and CD155 in HCC cell lines treated with meloxicam and further explore its possible mechanism. In vivo experiments were applied to verify the effect of meloxicam combined with anti-PD1 therapy on HCC tumor growth in mice. Results. Meloxicam can significantly inhibit the proliferation, invasion, and migration of HCC cells. The TISIDB database indicated that the COX2 was strongly associated with immunoinhibitors and immunostimulators. Meloxicam upregulated the level of PD-L1 in HCC cell lines and animal models. In terms of mechanism, meloxicam inhibited microRNA-200, thereby upregulating PD-L1. In vitro experiments showed that both meloxicam and anti-PD1 had inhibitory effects on the growth of HCC tumors. Compared with meloxicam and anti-PD1 alone, the combination therapy showed stronger antitumor properties. Immunohistochemical analysis confirmed that meloxicam enhanced the antitumor immune activity in the tumor microenvironment. Conclusion. Our study showed meloxicam inhibited HCC progression and enhanced the sensitivity of immunotherapy via the microRNA-200/PD-L1 pathway.
Collapse
|
85
|
Song S, Wen F, Gu S, Gu P, Huang W, Ruan S, Chen X, Zhou J, Li Y, Liu J, Shu P. Network Pharmacology Study and Experimental Validation of Yiqi Huayu Decoction Inducing Ferroptosis in Gastric Cancer. Front Oncol 2022; 12:820059. [PMID: 35237519 PMCID: PMC8883049 DOI: 10.3389/fonc.2022.820059] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2021] [Accepted: 01/17/2022] [Indexed: 12/24/2022] Open
Abstract
ObjectiveThis study aimed to identify the mechanism of Yiqi Huayu Decoction (YQHY) induced ferroptosis in gastric cancer (GC) by using network pharmacology and experimental validation.MethodsThe targets of YQHY, ferroptosis-related targets, and targets related to GC were derived from databases. Following the protein–protein interaction (PPI) network, the hub targets for YQHY induced ferroptosis in GC were identified. Furthermore, gene ontology (GO) and Kyoto Encyclopedia of Genes and Genomes (KEGG) enrichment were used to analyze the hub targets from a macro perspective. We verified the hub targets by molecular docking, GEPIA, HPA, and the cBioPortal database. Finally, we performed cell viability assays, quantitative real-time polymerase chain reaction (qRT-PCR), western blotting, lipid peroxidation, and GSH assays to explore the mechanism of YQHY induced ferroptosis in GC.ResultsWe identified the main active compounds and hub targets: Quercetin, DIBP, DBP, Mipax, Phaseol and TP53, ATM, SMAD4, PTGS2, and ACSL4. KEGG enrichment analyses indicated that the JAK2-STAT3 signaling pathway may be a significant pathway. Molecular docking results showed that the main active compounds had a good binding activity with the hub targets. The experimental results proved that YQHY could induce ferroptosis in AGS by increasing the MDA content and reducing the GSH content. qRT–PCR and Western blot results showed that YQHY can induce ferroptosis in GC by affecting the JAK2-STAT3 pathway and the expression of ACSL4.ConclusionsThis study indicated that YQHY can induce ferroptosis in GC by affecting the JAK2–STAT3 pathway and the expression of ACSL4, and induction of ferroptosis may be one of the possible mechanisms of YQHY’s anti-recurrence and metastasis of GC.
Collapse
Affiliation(s)
- Siyuan Song
- Department of Medical Oncology, Affiliated Hospital of Nanjing University of Chinese Medicine, Nanjing, China
- The First College for Clinical Medicine, Nanjing University of Chinese Medicine, Nanjing, China
- Department of Medical Oncology, Jiangsu Provincial Hospital of Chinese Medicine, Nanjing, China
| | - Fang Wen
- The First College for Clinical Medicine, Nanjing University of Chinese Medicine, Nanjing, China
| | - Suping Gu
- The First College for Clinical Medicine, Nanjing University of Chinese Medicine, Nanjing, China
| | - Peixin Gu
- The First College for Clinical Medicine, Nanjing University of Chinese Medicine, Nanjing, China
| | - Wenjie Huang
- Department of Medical Oncology, Affiliated Hospital of Nanjing University of Chinese Medicine, Nanjing, China
- The First College for Clinical Medicine, Nanjing University of Chinese Medicine, Nanjing, China
- Department of Medical Oncology, Jiangsu Provincial Hospital of Chinese Medicine, Nanjing, China
| | - Shuai Ruan
- Department of Medical Oncology, Affiliated Hospital of Nanjing University of Chinese Medicine, Nanjing, China
- The First College for Clinical Medicine, Nanjing University of Chinese Medicine, Nanjing, China
- Department of Medical Oncology, Jiangsu Provincial Hospital of Chinese Medicine, Nanjing, China
| | - Xiaoxue Chen
- Department of Medical Oncology, Affiliated Hospital of Nanjing University of Chinese Medicine, Nanjing, China
- The First College for Clinical Medicine, Nanjing University of Chinese Medicine, Nanjing, China
- Department of Medical Oncology, Jiangsu Provincial Hospital of Chinese Medicine, Nanjing, China
| | - Jiayu Zhou
- Department of Medical Oncology, Affiliated Hospital of Nanjing University of Chinese Medicine, Nanjing, China
- The First College for Clinical Medicine, Nanjing University of Chinese Medicine, Nanjing, China
- Department of Medical Oncology, Jiangsu Provincial Hospital of Chinese Medicine, Nanjing, China
| | - Ye Li
- Department of Medical Oncology, Affiliated Hospital of Nanjing University of Chinese Medicine, Nanjing, China
- The First College for Clinical Medicine, Nanjing University of Chinese Medicine, Nanjing, China
- Department of Medical Oncology, Jiangsu Provincial Hospital of Chinese Medicine, Nanjing, China
| | - Jiatong Liu
- The First College for Clinical Medicine, Nanjing University of Chinese Medicine, Nanjing, China
| | - Peng Shu
- Department of Medical Oncology, Affiliated Hospital of Nanjing University of Chinese Medicine, Nanjing, China
- The First College for Clinical Medicine, Nanjing University of Chinese Medicine, Nanjing, China
- Department of Medical Oncology, Jiangsu Provincial Hospital of Chinese Medicine, Nanjing, China
- *Correspondence: Peng Shu,
| |
Collapse
|
86
|
Usui N, Togawa S, Sumi T, Kobayashi Y, Koyama Y, Nakamura Y, Kondo M, Shinoda K, Kobayashi H, Shimada S. Si-Based Hydrogen-Producing Nanoagent Protects Fetuses From Miscarriage Caused by Mother-to-Child Transmission. FRONTIERS IN MEDICAL TECHNOLOGY 2022; 3:665506. [PMID: 35047922 PMCID: PMC8757766 DOI: 10.3389/fmedt.2021.665506] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2021] [Accepted: 04/06/2021] [Indexed: 12/24/2022] Open
Abstract
Mother-to-child transmission of viruses and bacteria increases the risk of miscarriage and various diseases in children. Such transmissions can result in infections and diseases in infants or the induction of an inflammatory immune response through the placenta. Recently, we developed a silicon (Si)-based hydrogen-producing nanoagent (Si-based agent) that continuously and effectively produces hydrogen in the body. Since medical hydrogen has antioxidative, anti-inflammatory, antiallergic, and antiapoptotic effects, we investigated the effects of our Si-based agent on mother-to-child transmission, with a focus on the rate of miscarriage. In pregnant mice fed a diet containing the Si-based agent, lipopolysaccharide (LPS)-induced miscarriage due to mother-to-child transmission was reduced and inflammation and neutrophil infiltration in the placenta were suppressed. We also found that the Si-based agent suppressed IL-6 expression in the placenta and induced the expression of antioxidant and antiapoptotic genes, such as Hmox1 and Ptgs2. The observed anti-inflammatory effects of the Si-based agent suggest that it may be an effective preventative or therapeutic drug for miscarriage or threatened miscarriage during pregnancy by suppressing maternal inflammation caused by bacterial and viral infections.
Collapse
Affiliation(s)
- Noriyoshi Usui
- Department of Neuroscience and Cell Biology, Graduate School of Medicine, Osaka University, Suita, Japan.,United Graduate School of Child Development, Osaka University, Suita, Japan.,Global Center for Medical Engineering and Informatics, Osaka University, Suita, Japan.,Addiction Research Unit, Osaka Psychiatric Research Center, Osaka Psychiatric Medical Center, Osaka, Japan
| | - Shogo Togawa
- Department of Neuroscience and Cell Biology, Graduate School of Medicine, Osaka University, Suita, Japan.,Division of Neuroanatomy, Department of Neuroscience, Yamaguchi University Graduate School of Medicine, Yamaguchi, Japan
| | - Takuya Sumi
- Department of Neuroscience and Cell Biology, Graduate School of Medicine, Osaka University, Suita, Japan.,Department of Cell Biology, Graduate School of Medicine, Osaka University, Suita, Japan
| | - Yuki Kobayashi
- Institute of Scientific and Industrial Research, Osaka University, Ibaraki, Japan
| | - Yoshihisa Koyama
- Department of Neuroscience and Cell Biology, Graduate School of Medicine, Osaka University, Suita, Japan.,Addiction Research Unit, Osaka Psychiatric Research Center, Osaka Psychiatric Medical Center, Osaka, Japan
| | - Yukiko Nakamura
- Department of Neuroscience and Cell Biology, Graduate School of Medicine, Osaka University, Suita, Japan.,Addiction Research Unit, Osaka Psychiatric Research Center, Osaka Psychiatric Medical Center, Osaka, Japan
| | - Makoto Kondo
- Department of Neuroscience and Cell Biology, Graduate School of Medicine, Osaka University, Suita, Japan.,Addiction Research Unit, Osaka Psychiatric Research Center, Osaka Psychiatric Medical Center, Osaka, Japan
| | - Koh Shinoda
- Division of Neuroanatomy, Department of Neuroscience, Yamaguchi University Graduate School of Medicine, Yamaguchi, Japan
| | - Hikaru Kobayashi
- Institute of Scientific and Industrial Research, Osaka University, Ibaraki, Japan
| | - Shoichi Shimada
- Department of Neuroscience and Cell Biology, Graduate School of Medicine, Osaka University, Suita, Japan.,United Graduate School of Child Development, Osaka University, Suita, Japan.,Addiction Research Unit, Osaka Psychiatric Research Center, Osaka Psychiatric Medical Center, Osaka, Japan
| |
Collapse
|
87
|
Mirzaie Z, Bastani A, Haji-Aghamohammadi AA, Rashidi Nooshabadi M, Ahadinezhad B, Khadem Haghighian H. Effects of Ellagic Acid on Oxidative Stress Index, Inflammatory Markers and Quality of Life in Patients With Irritable Bowel Syndrome: Randomized Double-blind Clinical Trial. Clin Nutr Res 2022; 11:98-109. [PMID: 35558999 PMCID: PMC9065395 DOI: 10.7762/cnr.2022.11.2.98] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2022] [Revised: 04/19/2022] [Accepted: 04/20/2022] [Indexed: 11/24/2022] Open
Abstract
Irritable bowel syndrome (IBS) is a common disorder that affects the large intestine. Oxidative stress and inflammation play a major role in IBS. Considering the antioxidant properties of ellagic acid (EA), this study was designed to evaluate the effect of EA on oxidative stress index, inflammatory markers, and quality of life in patients with IBS. This research was conducted as a randomized, double-blind, placebo-controlled clinical trial; 44 patients with IBS were recruited. Patients who met the inclusion criteria were randomly allocated to consume a capsule containing 180 mg of EA per day (n = 22) or a placebo (n = 22) for 8 weeks. Serum levels of total antioxidant capacity (TAC), malondialdehyde (MDA), C-reactive protein (CRP), and interleukin-6 (IL-6) were measured at the beginning and the end of the study. Also, quality of life was assessed using a self-report questionnaire for IBS patients (IBS-QOL). At the end of the study, we saw a significant decrease and increase in the MDA and TAC in the intervention group, respectively (p < 0.05). Also, EA consumption reduced CRP and IL-6 levels, and these changes were significant in comparison with placebo group changes (p < 0.05). The overall score of IBS-QOL significantly decreased, and quality of life was increased (p < 0.05), but there were no significant changes in the placebo group. According to these findings, receiving polyphenols, such as EA, may help maintain intestinal health by modulating inflammation and oxidative stress and ultimately improving the quality of life in IBS patients.
Collapse
Affiliation(s)
- Zahra Mirzaie
- Department of Nutrition, School of Health, Qazvin University of Medical Sciences, Qazvin 34197-59811, Iran
| | - Ali Bastani
- Department of Internal Medicine, Velayat Clinical Research Development Unit, Qazvin University of Medical Sciences, Qazvin 34197-59811, Iran
| | - Ali Akbar Haji-Aghamohammadi
- Department of Internal Medicine, Velayat Clinical Research Development Unit, Qazvin University of Medical Sciences, Qazvin 34197-59811, Iran
| | | | - Bahman Ahadinezhad
- Social Determinants of Health Research Center, Qazvin University of Medical Sciences, Qazvin 34197-59811, Iran
| | - Hossein Khadem Haghighian
- Department of Nutrition, School of Health, Qazvin University of Medical Sciences, Qazvin 34197-59811, Iran
- Metabolic Diseases Research Center, Research Institute for Prevention of Non-Communicable Diseases, Qazvin University of Medical Sciences, Qazvin 34197-59811, Iran
| |
Collapse
|
88
|
Biswas P, Datta HK, Dastidar P. Designing Coordination Polymers as Multi-drug-self-delivery System for Tuberculosis and Cancer Therapy: in vitro Viability and in vivo Toxicity Assessment. Biomater Sci 2022; 10:6201-6216. [PMID: 36097681 DOI: 10.1039/d2bm00752e] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
Abstract
A proof of the concept for designing multi-drug-delivery system suitable for self-drug-delivery is disclosed. Simple coordination chemistry was employed to anchor two kinds of drugs namely isoniazid (IZ – anti-tuberculosis),...
Collapse
Affiliation(s)
- Protap Biswas
- School of Chemical Sciences, Indian Association for the Cultivation of Science (IACS), 2A and 2B Raja S.C. Mullick Road, Jadavpur, Kolkata-700032, West Bengal, India.
| | - Hemanta Kumar Datta
- School of Chemical Sciences, Indian Association for the Cultivation of Science (IACS), 2A and 2B Raja S.C. Mullick Road, Jadavpur, Kolkata-700032, West Bengal, India.
| | - Parthasarathi Dastidar
- School of Chemical Sciences, Indian Association for the Cultivation of Science (IACS), 2A and 2B Raja S.C. Mullick Road, Jadavpur, Kolkata-700032, West Bengal, India.
| |
Collapse
|
89
|
Astaxanthin intake alleviates gouty arthritis in patients and rats by modulating the levels of various inflammatory markers. J Funct Foods 2021. [DOI: 10.1016/j.jff.2021.104823] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022] Open
|
90
|
Termer M, Carola C, Salazar A, Keck CM, Hemberger J, von Hagen J. Activity-Guided Characterization of COX-2 Inhibitory Compounds in Waltheria indica L. Extracts. Molecules 2021; 26:molecules26237240. [PMID: 34885813 PMCID: PMC8658768 DOI: 10.3390/molecules26237240] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2021] [Revised: 11/23/2021] [Accepted: 11/26/2021] [Indexed: 11/16/2022] Open
Abstract
Inflammation is the body's response to infection or tissue injury in order to restore and maintain homeostasis. Prostaglandin E2 (PGE-2) derived from arachidonic acid (AA), via up-regulation of cyclooxygenase-2 (COX-2), is a key mediator of inflammation and can also be induced by several other factors including stress, chromosomal aberration, or environmental factors. Targeting prostaglandin production by inhibiting COX-2 is hence relevant for the successful resolution of inflammation. Waltheria indica L. is a traditional medicinal plant whose extracts have demonstrated COX-2 inhibitory properties. However, the compounds responsible for the activity remained unknown. For the preparation of extracts with effective anti-inflammatory properties, characterization of these substances is vital. In this work, we aimed to address this issue by characterizing the substances responsible for the COX-2 inhibitory activity in the extracts and generating prediction models to quantify the COX-2 inhibitory activity without biological testing. For this purpose, an extract was separated into fractions by means of centrifugal partition chromatography (CPC). The inhibitory potential of the fractions and extracts against the COX-2 enzyme was determined using a fluorometric COX-2 inhibition assay. The characterizations of compounds in the fractions with the highest COX-2 inhibitory activity were conducted by high resolution mass spectrometry (HPLC-MS/MS). It was found that these fractions contain alpha-linolenic acid, linoleic acid and oleic acid, identified and reported for the first time in Waltheria indica leaf extracts. After analyzing their contents in different Waltheria indica extracts, it could be demonstrated that these fatty acids are responsible for up to 41% of the COX-2 inhibition observed with Waltheria indica extract. Additional quantification of secondary metabolites in the extract fractions revealed that substances from the group of steroidal saponins and triterpenoid saponins also contribute to the COX-2 inhibitory activity. Based on the content of compounds contributing to COX-2 inhibition, two mathematical models were successfully developed, both of which had a root mean square error (RMSE) = 1.6% COX-2 inhibitory activity, demonstrating a high correspondence between predicted versus observed values. The results of the predictive models further suggested that the compounds contribute to COX-2 inhibition in the order linoleic acid > alpha linolenic acid > steroidal saponins > triterpenoid saponins. The characterization of substances contributing to COX-2 inhibition in this study enables a more targeted development of extraction processes to obtain Waltheria indica extracts with superior anti-inflammatory properties.
Collapse
Affiliation(s)
- Michael Termer
- Department of Pharmaceutics and Biopharmaceutics, Philipps-University of Marburg, Robert-Koch-Str. 4, 35032 Marburg, Germany;
- Correspondence:
| | - Christophe Carola
- Merck KGaA, Frankfurterstr. 250, 64293 Darmstadt, Germany; (C.C.); (A.S.); (J.v.H.)
| | - Andrew Salazar
- Merck KGaA, Frankfurterstr. 250, 64293 Darmstadt, Germany; (C.C.); (A.S.); (J.v.H.)
| | - Cornelia M. Keck
- Department of Pharmaceutics and Biopharmaceutics, Philipps-University of Marburg, Robert-Koch-Str. 4, 35032 Marburg, Germany;
| | - Juergen Hemberger
- Department of Life Science Engineering, Institute for Biochemical Engineering & Analytics, University of Applied Sciences, Wiesenstr. 14, 35390 Giessen, Germany;
| | - Joerg von Hagen
- Merck KGaA, Frankfurterstr. 250, 64293 Darmstadt, Germany; (C.C.); (A.S.); (J.v.H.)
- Department of Life Science Engineering, Institute for Biochemical Engineering & Analytics, University of Applied Sciences, Wiesenstr. 14, 35390 Giessen, Germany;
| |
Collapse
|
91
|
Pla-Pagà L, Valls RM, Pedret A, Calderón-Pérez L, Llauradó E, Companys J, Domenech-Coca C, Canela N, Del Bas JM, Caimari A, Puiggròs F, Mi C, Arola L, Solà R. Effect of the consumption of hesperidin in orange juice on the transcriptomic profile of subjects with elevated blood pressure and stage 1 hypertension: A randomized controlled trial (CITRUS study). Clin Nutr 2021; 40:5812-5822. [PMID: 34800819 DOI: 10.1016/j.clnu.2021.10.009] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2021] [Revised: 09/21/2021] [Accepted: 10/14/2021] [Indexed: 12/19/2022]
Abstract
SCOPE Hesperidin exerts cardiovascular beneficial effects, but its mechanisms of action remain undefined. In a previous study we demonstrated that a single dose and a 12-week treatment of hesperidin decreased systolic blood pressure. The aim of this study was to ascertain the action mechanisms of hesperidin consumption in subjects with elevated blood pressure or with stage 1 hypertension, by determining their transcriptomic profile after a single dose or a 12-week treatment. METHODS AND RESULTS For transcriptomic analysis, peripheral blood mononuclear cells were obtained from 37 subjects with elevated blood pressure and stage 1 hypertension from CITRUS study who were randomized to receive for 12 weeks: control drink (CD; n = 11), OJ (containing 345 mg of hesperidin; n = 15) or EOJ (containing 600 mg of hesperidin; n = 11). Before starting the 12-weeks treatment, a single dose study with a 6 h of follow-up in each group was performed. After the single dose consumption, EOJ versus OJ, downregulated DHRS9 gene which is related with insulin resistance. Compared to CD, 12-week treatment of EOJ downregulated 6 proinflammatory genes while after OJ consumption only 1 proinflammatory gene was downregulated. Moreover, 12-week treatment of EOJ versus OJ, downregulated acute coronary syndrome gene related (SELENBP1). CONCLUSION A single dose consumption of EOJ could protect from insulin resistance. Moreover, EOJ decrease the expression of proinflammatory genes after 12-week treatment providing a possible mechanism of action on inflammation pathway.
Collapse
Affiliation(s)
- Laura Pla-Pagà
- Eurecat, Centre Tecnològic de Catalunya, Unitat de Nutrició i Salut, Reus, Spain; Universitat Rovira i Virgili, Facultat de Medicina i Ciències de la Salut, Functional Nutrition, Oxidation and Cardiovascular Disease Group (NFOC-Salut), Reus, Spain
| | - Rosa M Valls
- Eurecat, Centre Tecnològic de Catalunya, Unitat de Nutrició i Salut, Reus, Spain; Universitat Rovira i Virgili, Facultat de Medicina i Ciències de la Salut, Functional Nutrition, Oxidation and Cardiovascular Disease Group (NFOC-Salut), Reus, Spain.
| | - Anna Pedret
- Eurecat, Centre Tecnològic de Catalunya, Unitat de Nutrició i Salut, Reus, Spain; Universitat Rovira i Virgili, Facultat de Medicina i Ciències de la Salut, Functional Nutrition, Oxidation and Cardiovascular Disease Group (NFOC-Salut), Reus, Spain.
| | - Lorena Calderón-Pérez
- Eurecat, Centre Tecnològic de Catalunya, Unitat de Nutrició i Salut, Reus, Spain; Universitat Rovira i Virgili, Facultat de Medicina i Ciències de la Salut, Functional Nutrition, Oxidation and Cardiovascular Disease Group (NFOC-Salut), Reus, Spain
| | - Elisabet Llauradó
- Universitat Rovira i Virgili, Facultat de Medicina i Ciències de la Salut, Functional Nutrition, Oxidation and Cardiovascular Disease Group (NFOC-Salut), Reus, Spain
| | - Judit Companys
- Eurecat, Centre Tecnològic de Catalunya, Unitat de Nutrició i Salut, Reus, Spain; Universitat Rovira i Virgili, Facultat de Medicina i Ciències de la Salut, Functional Nutrition, Oxidation and Cardiovascular Disease Group (NFOC-Salut), Reus, Spain
| | | | - Nuria Canela
- Eurecat, Centre Tecnològic de Catalunya, Centre for Omic Sciences, Joint Unit Universitat Rovira i Virgili-EURECAT, Reus, Spain
| | - Josep M Del Bas
- Eurecat, Centre Tecnològic de Catalunya, Unitat de Nutrició i Salut, Reus, Spain
| | - Antoni Caimari
- Eurecat, Centre Tecnològic de Catalunya, Unitat de Nutrició i Salut, Reus, Spain
| | - Francesc Puiggròs
- Eurecat, Centre Tecnològic de Catalunya, Centre for Omic Sciences, Joint Unit Universitat Rovira i Virgili-EURECAT, Reus, Spain
| | - Covas Mi
- Nutritional Projects Assessment (Nuproas.es), Barcelona, Spain
| | - Lluís Arola
- Eurecat, Centre Tecnològic de Catalunya, Centre for Omic Sciences, Joint Unit Universitat Rovira i Virgili-EURECAT, Reus, Spain
| | - Rosa Solà
- Universitat Rovira i Virgili, Facultat de Medicina i Ciències de la Salut, Functional Nutrition, Oxidation and Cardiovascular Disease Group (NFOC-Salut), Reus, Spain; Hospital Universitari Sant Joan de Reus, Reus, Spain
| |
Collapse
|
92
|
Regulation of inflammation and COX-2 gene expression in benzo (a) pyrene induced lung carcinogenesis in mice by all trans retinoic acid (ATRA). Life Sci 2021; 285:119967. [PMID: 34543639 DOI: 10.1016/j.lfs.2021.119967] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2021] [Revised: 09/05/2021] [Accepted: 09/13/2021] [Indexed: 11/21/2022]
Abstract
AIM Inflammation provides favourable microenvironment for cancer development. An enhanced COX-2 gene expression is a key inflammatory mediator of cancers and the drug that inhibits it, helps to manage cancer effectively and increases survival rate. The objective is to analyse the inflammatory changes and COX-2 gene expression in benzo (a) pyrene induced mice and to evaluate the regulatory effect of all trans retinoic acid. MATERIALS AND METHODS The body and organ weights were recorded in B(a)P induced mice. The haematological parameters and serum inflammatory markers of carcinogenesis were tested. The H & E stained liver and lung tissues were examined for histopathologic changes. The COX-2 gene expression was analysed by RT-PCR and qPCR in lung and liver. KEY FINDINGS The decreased body weight, increased organ weights and the damages in liver and lung were observed in B(a)P induced mice and were prevented significantly upon ATRA treatment. The lowered Hb, RBC and lymphocytes and an enhanced WBC, monocytes and neutrophils observed in B(a)P group were significantly reversed in treated group. A drastic increase in cancer associated inflammatory markers observed in B(a)P induced mice were significantly (P ≤ 0.001) reduced in treated mice. The RT-PCR product density of COX-2 gene was very high in B(a)P group (lung-0.43 ± 0.06; liver-0.39 ± 0.04) significantly lower in treated group (lung-0.12 ± 0.03; liver-0.08 ± 0.03) with a significant difference in RQ values (B(a)P lung-18.46 ± 0.04, liver-12.46 ± 0.08; treated lung-5.93 ± 0.07, liver-2.92 ± 0.10). SIGNIFICANCE The ATRA has decreased the inflammatory condition with downregulation of COX-2 gene expression and thereby prevented carcinogenesis during early stage of B(a)P induced cancer development.
Collapse
|
93
|
Herrera-Vargas AK, García-Rodríguez E, Olea-Flores M, Mendoza-Catalán MA, Flores-Alfaro E, Navarro-Tito N. Pro-angiogenic activity and vasculogenic mimicry in the tumor microenvironment by leptin in cancer. Cytokine Growth Factor Rev 2021; 62:23-41. [PMID: 34736827 DOI: 10.1016/j.cytogfr.2021.10.006] [Citation(s) in RCA: 40] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2021] [Revised: 10/18/2021] [Accepted: 10/18/2021] [Indexed: 12/11/2022]
Abstract
The acquired ability to induce the formation of a functional vasculature is a hallmark of cancer. Blood vessels in tumors are formed through various mechanisms, among the most important in cancer biology, angiogenesis, and vasculogenic mimicry have been described. Leptin is one of the main adipokines secreted by adipocytes in normal breast tissue and the tumor microenvironment. Here, we provide information on the relationship between leptin and the development of angiogenesis and vasculogenic mimicry in different types of cancer. Here, we report that leptin activates different pathways such as JAK-STAT3, MAPK/ERK, PKC, JNK, p38, and PI3K-Akt to induce the expression of various angiogenic factors and vasculogenic mimicry. In vivo models, leptin induces blood vessel formation through the PI3K-Akt-mTOR pathway. Interestingly, the relationship between leptin and vasculogenic mimicry was more significant in breast cancer. The information obtained suggests that leptin could be playing an essential role in tumor survival and metastasis through the induction of vascular mechanisms such as angiogenesis and vasculogenic mimicry; thus, leptin-induced pathways could be suggested as a promising therapeutic target.
Collapse
Affiliation(s)
- Ana K Herrera-Vargas
- Laboratorio de Biología Celular del Cáncer, Facultad de Ciencias Químico Biológicas, Universidad Autónoma de Guerrero, Av. Lázaro Cárdenas s/n, Chilpancingo, GRO 39090, Mexico.
| | - Eduardo García-Rodríguez
- Laboratorio de Biología Celular del Cáncer, Facultad de Ciencias Químico Biológicas, Universidad Autónoma de Guerrero, Av. Lázaro Cárdenas s/n, Chilpancingo, GRO 39090, Mexico.
| | - Monserrat Olea-Flores
- Laboratorio de Biología Celular del Cáncer, Facultad de Ciencias Químico Biológicas, Universidad Autónoma de Guerrero, Av. Lázaro Cárdenas s/n, Chilpancingo, GRO 39090, Mexico.
| | - Miguel A Mendoza-Catalán
- Laboratorio de Biomedicina Molecular, Facultad de Ciencias Químico Biológicas, Universidad Autónoma de Guerrero, Chilpancingo, GRO, 39090, Mexico.
| | - Eugenia Flores-Alfaro
- Laboratorio de Epidemiología Clínica y Molecular, Facultad de Ciencias Químico-Biológicas, Universidad Autónoma de Guerrero, Chilpancingo, GRO 39087, Mexico.
| | - Napoleón Navarro-Tito
- Laboratorio de Biología Celular del Cáncer, Facultad de Ciencias Químico Biológicas, Universidad Autónoma de Guerrero, Av. Lázaro Cárdenas s/n, Chilpancingo, GRO 39090, Mexico.
| |
Collapse
|
94
|
Ramirez MF, Rangel FP, Cata JP. Perioperative pain, analgesics and cancer-related outcomes: where do we stand? Pain Manag 2021; 12:229-242. [PMID: 34636651 DOI: 10.2217/pmt-2021-0070] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022] Open
Abstract
Cancer-related pain is one of the most common and debilitating symptoms among cancer patients. Undertreated cancer-related pain interferes with daily activities and increases morbidity and mortality. While opioids continue to play an essential role in treating moderate to severe cancer-related pain, they are associated with many adverse effects including misuse. While preclinical and retrospective studies have shown a negative association between opioid use and cancer outcomes, randomized control trials demonstrate that opioid use does not influence cancer recurrence. Additionally, analgesics and adjuvants used for perioperatively or chronic pain control are unlikely to improve oncological outcomes. This article focuses on the pharmacological management of cancer-related pain and offers an overview regarding the use of these medications perioperatively and the cancer outcomes.
Collapse
Affiliation(s)
- Maria F Ramirez
- Department of Anesthesiology & Perioperative Medicine, The University of Texas MD Anderson Cancer Center, Houston, TX 77005, USA.,Anesthesiology & Surgical Oncology Research Group, Houston, TX, USA
| | | | - Juan P Cata
- Department of Anesthesiology & Perioperative Medicine, The University of Texas MD Anderson Cancer Center, Houston, TX 77005, USA.,Anesthesiology & Surgical Oncology Research Group, Houston, TX, USA
| |
Collapse
|
95
|
Othman MS, Al-Bagawi AH, Obeidat ST, Fareid MA, Habotta OA, Moneim AEA. Antitumor activity of zinc nanoparticles synthesized with berberine on human epithelial colorectal adenocarcinoma (Caco-2) cells through acting on Cox-2/NF-kB and p53 pathways. Anticancer Agents Med Chem 2021; 22:2002-2010. [PMID: 34607550 DOI: 10.2174/1871520621666211004115839] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2021] [Revised: 07/11/2021] [Accepted: 08/05/2021] [Indexed: 11/22/2022]
Abstract
BACKGROUND Drawbacks and side effects of currently available therapies to colorectal cancer (CRC) devoted the researchers to search for new therapeutic strategies. OBJECTIVE This study was designed to investigate the effects of zinc nanoparticles biosynthesized with berberine (ZnNPs-BER) on Caco-2 cells compared to 5-Fluorouracil (5-FU) and explore the possible underlying pathways. METHODS Caco-2 and Vero cells were treated with 5-FU, BER, or ZnNPs-BER for 24 h. Cell viability was measured by MTT assay. Oxidative stress and apoptotic markers and cell cycle were determined. Additionally, Cox-2 and NF-kB levels were also measured. RESULTS The IC50 of 5-FU, BER, and ZnNPs-BER on Caco-2 cells were 34.65 µM, 19.86 µg/ml and 10.49 µg/ml, respectively by MTT assay. The IC50 value for 5-FU in Vero cells was 21.7 μg/ml, however, BER and BER-ZnNPs treatment showed non-toxic effects to the Vero cells. Further, ZnNPs-BER exerted significant induction of ROS besides exhaustion of the antioxidant capacity of tumor cells indicated by declined GSH and elevated NO and MDA contents. Marked increments in levels of Bax and caspase-3 were detected together with declines in Bcl-2 levels in Caco-2 cells submitted to BER-ZnNPs therapy. On the molecular basis, upregulation in mRNA levels of pro-apoptotic genes (Bax, caspase-3, and tumor suppressor gene p53) with downregulation in the antiapoptotic gene (Bcl-2) were observed in ZnNPs-BER treated Caco-2 cells. Furthermore, ZnNPs-BER showed more pronounced effects on apoptosis increased cell percentage in the S and subG1 phases. In addition, green synthesis of ZnNPs with BER showed notable induction of Cox2 and NF-kB in Caco-2 cells. CONCLUSION Therefore, the antitumor potential of ZnNPs-BER in colon cancer cells may be endorsed for induction of oxidative stress, inflammation, and apoptotic changes in tumor cells. Our study documents the new therapeutic potential of Zn nanoparticles conjugated with BER, as a new option for combined chemotherapy.
Collapse
Affiliation(s)
- Mohamed S Othman
- Basic Sciences Department, Deanship of Preparatory Year, University of Ha'il, Hail. Saudi Arabia
| | - Amal H Al-Bagawi
- Chemistry Department, Faculty of Science, University of Ha'il, Hail. Saudi Arabia
| | - Sofian T Obeidat
- Basic Sciences Department, Deanship of Preparatory Year, University of Ha'il, Hail. Saudi Arabia
| | - Mohamed A Fareid
- Basic Sciences Department, Deanship of Preparatory Year, University of Ha'il, Hail. Saudi Arabia
| | - Ola A Habotta
- Department of Forensic Medicine and Toxicology, Faculty of Veterinary Medicine, Mansoura University, Mansoura. Egypt
| | - Ahmed E Abdel Moneim
- Zoology and Entomology Department, Faculty of Science, Helwan University, Cairo. Egypt
| |
Collapse
|
96
|
Ávila-Román J, García-Gil S, Rodríguez-Luna A, Motilva V, Talero E. Anti-Inflammatory and Anticancer Effects of Microalgal Carotenoids. Mar Drugs 2021; 19:531. [PMID: 34677429 PMCID: PMC8539290 DOI: 10.3390/md19100531] [Citation(s) in RCA: 52] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2021] [Revised: 09/19/2021] [Accepted: 09/20/2021] [Indexed: 12/12/2022] Open
Abstract
Acute inflammation is a key component of the immune system's response to pathogens, toxic agents, or tissue injury, involving the stimulation of defense mechanisms aimed to removing pathogenic factors and restoring tissue homeostasis. However, uncontrolled acute inflammatory response may lead to chronic inflammation, which is involved in the development of many diseases, including cancer. Nowadays, the need to find new potential therapeutic compounds has raised the worldwide scientific interest to study the marine environment. Specifically, microalgae are considered rich sources of bioactive molecules, such as carotenoids, which are natural isoprenoid pigments with important beneficial effects for health due to their biological activities. Carotenoids are essential nutrients for mammals, but they are unable to synthesize them; instead, a dietary intake of these compounds is required. Carotenoids are classified as carotenes (hydrocarbon carotenoids), such as α- and β-carotene, and xanthophylls (oxygenate derivatives) including zeaxanthin, astaxanthin, fucoxanthin, lutein, α- and β-cryptoxanthin, and canthaxanthin. This review summarizes the present up-to-date knowledge of the anti-inflammatory and anticancer activities of microalgal carotenoids both in vitro and in vivo, as well as the latest status of human studies for their potential use in prevention and treatment of inflammatory diseases and cancer.
Collapse
Affiliation(s)
- Javier Ávila-Román
- Department of Biochemistry and Biotechnology, Universitat Rovira i Virgili, 43007 Tarragona, Spain
| | - Sara García-Gil
- Department of Pharmacology, Universidad de Sevilla, 41012 Seville, Spain; (S.G.-G.); (A.R.-L.); (V.M.)
| | - Azahara Rodríguez-Luna
- Department of Pharmacology, Universidad de Sevilla, 41012 Seville, Spain; (S.G.-G.); (A.R.-L.); (V.M.)
| | - Virginia Motilva
- Department of Pharmacology, Universidad de Sevilla, 41012 Seville, Spain; (S.G.-G.); (A.R.-L.); (V.M.)
| | - Elena Talero
- Department of Pharmacology, Universidad de Sevilla, 41012 Seville, Spain; (S.G.-G.); (A.R.-L.); (V.M.)
| |
Collapse
|
97
|
Vinhas A, Gonçalves AI, Rodrigues MT, Gomes ME. Human tendon-derived cell sheets created by magnetic force-based tissue engineering hold tenogenic and immunomodulatory potential. Acta Biomater 2021; 131:236-247. [PMID: 34192569 DOI: 10.1016/j.actbio.2021.06.036] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2021] [Revised: 06/14/2021] [Accepted: 06/22/2021] [Indexed: 02/07/2023]
Abstract
Cell sheet technology and magnetic based tissue engineering hold the potential to become instrumental in developing magnetically responsive living tissues analogues that can be potentially used both for modeling and therapeutical purposes. Cell sheet constructions more closely recreate physiological niches, through the preservation of contiguous cells and cell-ECM interactions, which assist the cellular guidance in regenerative processes. We herein propose to use magnetically assisted cell sheets (magCSs) constructed with human tendon-derived cells (hTDCs) and magnetic nanoparticles to study inflammation activity upon magCSs exposure to IL-1β, anticipating its added value for tendon disease modeling. Our results show that IL-1β induces an inflammatory profile in magCSs, supporting its in vitro use to enlighten inflammation mediated events in tendon cells. Moreover, the response of magCSs to IL-1β is modulated by pulsed electromagnetic field (PEMF) stimulation, favoring the expression of anti-inflammatory genes, which seems to be associated to MAPK(ERK1/2) pathway. The anti-inflammatory response to PEMF together with the immunomodulatory potential of magCSs opens new perspectives for their applicability on tendon regeneration that goes beyond advanced cell based modeling. STATEMENT OF SIGNIFICANCE: The combination of cell sheets and magnetic-based technologies holds promise as instrumental bio-instructive tools both for tendon disease modelling and for the development of magnetically responsive living tendon substitutes. We have previously shown that remote actuation of a pulsed electromagnetic field (PEMF) modulated the inflammatory response of IL-1β-treated human tendon-derived cell (hTDCs) monolayers. As magnetic cell sheets (magCSs) technologies enable improved cellular organization and matrix deposition, these constructions could better recapitulate tendon niches. In this work, we aimed to apply magCSs technologies to study hTDCs responses in inflammatory environments. Overall results show that PEMF-stimulated-magCSs hold evidence for immunomodulatory properties and to become a living tendon model envisioning tendon regenerative therapies.
Collapse
Affiliation(s)
- Adriana Vinhas
- 3B's Research Group, I3Bs - Research Institute on Biomaterials, Biodegradables and Biomimetics, University of Minho, Headquarters of the European Institute of Excellence on Tissue Engineering and Regenerative Medicine, Avepark - Zona Industrial da Gandra, 4805-017 Barco, Guimarães, Portugal; ICVS/3B's - PT Government Associate Laboratory, Braga/Guimarães, Portugal
| | - Ana I Gonçalves
- 3B's Research Group, I3Bs - Research Institute on Biomaterials, Biodegradables and Biomimetics, University of Minho, Headquarters of the European Institute of Excellence on Tissue Engineering and Regenerative Medicine, Avepark - Zona Industrial da Gandra, 4805-017 Barco, Guimarães, Portugal; ICVS/3B's - PT Government Associate Laboratory, Braga/Guimarães, Portugal
| | - Márcia T Rodrigues
- 3B's Research Group, I3Bs - Research Institute on Biomaterials, Biodegradables and Biomimetics, University of Minho, Headquarters of the European Institute of Excellence on Tissue Engineering and Regenerative Medicine, Avepark - Zona Industrial da Gandra, 4805-017 Barco, Guimarães, Portugal; ICVS/3B's - PT Government Associate Laboratory, Braga/Guimarães, Portugal.
| | - Manuela E Gomes
- 3B's Research Group, I3Bs - Research Institute on Biomaterials, Biodegradables and Biomimetics, University of Minho, Headquarters of the European Institute of Excellence on Tissue Engineering and Regenerative Medicine, Avepark - Zona Industrial da Gandra, 4805-017 Barco, Guimarães, Portugal; ICVS/3B's - PT Government Associate Laboratory, Braga/Guimarães, Portugal.
| |
Collapse
|
98
|
Cardoso C, Matos J, Gomes‐Bispo A, Afonso C, Motta C, Castanheira I, Prates JAM, Bandarra NM. Antioxidant and anti‐inflammatory activities of ethyl acetate extracts of chub mackerel (
Scomber colias
): a thorough seasonal evaluation. Int J Food Sci Technol 2021. [DOI: 10.1111/ijfs.15185] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Affiliation(s)
- Carlos Cardoso
- Division of Aquaculture, Upgrading, and Bioprospection (DivAV) Portuguese Institute for the Sea and Atmosphere (IPMA, IP) Avenida Alfredo Magalhães Ramalho, 6 Algés 1495‐165 Portugal
- CIIMAR Interdisciplinary Centre of Marine and Environmental Research University of Porto Rua dos Bragas 289 Porto 4050‐123 Portugal
| | - Joana Matos
- Division of Aquaculture, Upgrading, and Bioprospection (DivAV) Portuguese Institute for the Sea and Atmosphere (IPMA, IP) Avenida Alfredo Magalhães Ramalho, 6 Algés 1495‐165 Portugal
- Faculdade de Ciências da Universidade de Lisboa Campo Grande, 16 Lisboa 1749‐016 Portugal
| | - Ana Gomes‐Bispo
- Division of Aquaculture, Upgrading, and Bioprospection (DivAV) Portuguese Institute for the Sea and Atmosphere (IPMA, IP) Avenida Alfredo Magalhães Ramalho, 6 Algés 1495‐165 Portugal
- CIIMAR Interdisciplinary Centre of Marine and Environmental Research University of Porto Rua dos Bragas 289 Porto 4050‐123 Portugal
| | - Cláudia Afonso
- Division of Aquaculture, Upgrading, and Bioprospection (DivAV) Portuguese Institute for the Sea and Atmosphere (IPMA, IP) Avenida Alfredo Magalhães Ramalho, 6 Algés 1495‐165 Portugal
- CIIMAR Interdisciplinary Centre of Marine and Environmental Research University of Porto Rua dos Bragas 289 Porto 4050‐123 Portugal
| | - Carla Motta
- Instituto Nacional de Saúde Doutor Ricardo Jorge Av. Padre Cruz Lisbon 1600‐560 Portugal
| | - Isabel Castanheira
- Instituto Nacional de Saúde Doutor Ricardo Jorge Av. Padre Cruz Lisbon 1600‐560 Portugal
| | - José A. M. Prates
- Centro de Investigação Interdisciplinar em Sanidade Animal (CIISA) Faculdade de Medicina Veterinária Universidade de Lisboa Lisbon 1300‐477 Portugal
| | - Narcisa M. Bandarra
- Division of Aquaculture, Upgrading, and Bioprospection (DivAV) Portuguese Institute for the Sea and Atmosphere (IPMA, IP) Avenida Alfredo Magalhães Ramalho, 6 Algés 1495‐165 Portugal
- CIIMAR Interdisciplinary Centre of Marine and Environmental Research University of Porto Rua dos Bragas 289 Porto 4050‐123 Portugal
| |
Collapse
|
99
|
Luo Z, Ma Y, Di T, Ma B, Li H, An J, Wang Y, Zhang H. DNMT3B decreases extracellular matrix degradation and alleviates intervertebral disc degeneration through TRPA1 methylation to inhibit the COX2/YAP axis. Aging (Albany NY) 2021; 13:20258-20276. [PMID: 34428744 PMCID: PMC8436916 DOI: 10.18632/aging.203410] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2020] [Accepted: 02/08/2021] [Indexed: 01/14/2023]
Abstract
Intervertebral disc degeneration (IVDD) is a main cause of low back pain that is associated with extracellular matrix (ECM) degradation and inflammation. This study aims to investigate the role of DNMT3B and its regulatory mechanisms in IVDD. IVDD rat models were constructed followed by transfections with oe-DNMT3B or oe-YAP in order to explore the role of DNMT3B in the development of IVDD. After that transfection, nucleus pulposus (NP) cells were isolated and transfected with oe-DNMT3B, oe-TRPA1, si-YAP, oe-YAP or oe-COX2 in order to investigate the functions of DNMT3B in NP cells. DNMT3B was poorly expressed in IVDD tissues and NP cells whereas TRPA1, COX2, and YAP were highly expressed. The proliferation or apoptosis of NP cells was detected through CCK-8 assay or flow cytometry, respectively. Overexpression of DNMT3B promoted the proliferation of NP cells, inhibited their apoptosis, as well as increasing the expression of collagen II and aggrecan and decreasing expression of MMP3 and MMP9. Besides, DNMT3B suppressed inflammation and alleviated IVDD. Mechanistically, DNMT3B modified the TRPA1 promoter by methylation to inhibit the expression of COX2. Overexpression of COX2 promoted the apoptosis of NP cells and decreased the expression of YAP, which was reversed by upregulating DNMT3B. DNMT3B may promote the proliferation of NP cells and prevent their ECM degradation through the TRPA1/COX2/YAP axis, thereby alleviating IVDD in rats.
Collapse
Affiliation(s)
- Zhiqiang Luo
- Department of Orthopedics, Lanzhou University Second Hospital, Lanzhou 730030, P.R. China
| | - Yanchao Ma
- Department of Orthopedics, Lanzhou University Second Hospital, Lanzhou 730030, P.R. China
| | - Tianning Di
- Department of Orthopedics, Lanzhou University Second Hospital, Lanzhou 730030, P.R. China
| | - Bing Ma
- Department of Orthopedics, Lanzhou University Second Hospital, Lanzhou 730030, P.R. China
| | - Hongwei Li
- Department of Orthopedics, Lanzhou University Second Hospital, Lanzhou 730030, P.R. China
| | - Jiangdong An
- Department of Orthopedics, Lanzhou University Second Hospital, Lanzhou 730030, P.R. China
| | - Yonggang Wang
- Department of Orthopedics, Lanzhou University Second Hospital, Lanzhou 730030, P.R. China
| | - Haihong Zhang
- Department of Orthopedics, Lanzhou University Second Hospital, Lanzhou 730030, P.R. China
| |
Collapse
|
100
|
Tang B, Zhu J, Fang S, Wang Y, Vinothkumar R, Li M, Weng Q, Zheng L, Yang Y, Qiu R, Xu M, Zhao Z, Ji J. Pharmacological inhibition of MELK restricts ferroptosis and the inflammatory response in colitis and colitis-propelled carcinogenesis. Free Radic Biol Med 2021; 172:312-329. [PMID: 34144192 DOI: 10.1016/j.freeradbiomed.2021.06.012] [Citation(s) in RCA: 48] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/07/2021] [Accepted: 06/12/2021] [Indexed: 12/11/2022]
Abstract
INTRODUCTION Inflammatory bowel disease (IBD), including ulcerative colitis (UC) and Crohn's disease (CD), is a group of chronic recurrent and incurable gastrointestinal diseases with an unknown etiology that leads to a high risk of developing colitis-associated colorectal cancer (CRC). OBJECTIVES In this study, we measured the expression characteristics of MELK in IBD and CRC tissues and explored the regulatory effect of OTSSP167 (a MELK-selective inhibitor) on the mice models of colitis and colitis-associated carcinogenesis and analyzed the specific molecular mechanisms. METHODS DSS-induced colitis and colitis-associated carcinogenesis (CAC) model were treated with MELK inhibitor OTSSP167 then the fight against effect of OTSSP167 in the clinical symptoms of colitis and CAC was measured. In addition, underlying mechanism of OTSSP167 treatment in vitro and vivo including anti-ferroptosis and anti-inflammatory response effect was further explored. RESULTS We found that pharmacological inhibition of MELK was indicated to significantly alleviate the inflammatory response in mice with colitis, reduce intestinal damage, and effectively inhibit the occurrence and progression of colitis-propelled carcinogenesis, which was closely related to the regulation of gut microbial composition, and OTSSP167-mediated fecal microbiota transplantation effectively alleviated DSS-induced colitis. In addition, OTSSP167 treatment obviously inhibited ferroptosis in the intestinal tissue and suppressed macrophage infiltration and M1 polarization, which reduced the secretion of pro-inflammatory factors. Further exploration of the molecular mechanism revealed that OTSSP167 inhibited AKT/IKK/P65 and ERK/IKK/P65 signaling cascades both in vivo and in vitro, which may help alleviate intestinal inflammation and control the occurrence of cancer. CONCLUSION Our findings lay a theoretical foundation for the use of OTSSP167 as a treatment for IBD and its inhibition of the occurrence of colitis-associated carcinogenesis; additionally, MELK may be a potentially effective target molecule, thus providing more options for clinical treatment.
Collapse
Affiliation(s)
- Bufu Tang
- Key Laboratory of Imaging Diagnosis and Minimally Invasive Intervention Research, Lishui Hospital, School of Medicine, Zhejiang University, Lishui, 323000, China; Department of Radiology, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, 310016, China
| | - Jinyu Zhu
- Key Laboratory of Imaging Diagnosis and Minimally Invasive Intervention Research, Lishui Hospital, School of Medicine, Zhejiang University, Lishui, 323000, China; Department of Radiology, Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, China
| | - Shiji Fang
- Department of Radiology, The Fifth Affiliated Hospital of Wenzhou Medical University, Lishui, 323000, China
| | - Yajie Wang
- Key Laboratory of Imaging Diagnosis and Minimally Invasive Intervention Research, Lishui Hospital, School of Medicine, Zhejiang University, Lishui, 323000, China; Department of Radiology, The Fifth Affiliated Hospital of Wenzhou Medical University, Lishui, 323000, China
| | - Rajamanickam Vinothkumar
- Key Laboratory of Imaging Diagnosis and Minimally Invasive Intervention Research, Lishui Hospital, School of Medicine, Zhejiang University, Lishui, 323000, China
| | - Mengyao Li
- School of Basic Medical Sciences, Zhejiang University, Hangzhou, 310016, China
| | - Qiaoyou Weng
- Department of Radiology, The Fifth Affiliated Hospital of Wenzhou Medical University, Lishui, 323000, China
| | - Liyun Zheng
- Department of Radiology, The Fifth Affiliated Hospital of Wenzhou Medical University, Lishui, 323000, China
| | - Yang Yang
- Key Laboratory of Imaging Diagnosis and Minimally Invasive Intervention Research, Lishui Hospital, School of Medicine, Zhejiang University, Lishui, 323000, China; Department of Radiology, The Fifth Affiliated Hospital of Wenzhou Medical University, Lishui, 323000, China
| | - Rongfang Qiu
- Key Laboratory of Imaging Diagnosis and Minimally Invasive Intervention Research, Lishui Hospital, School of Medicine, Zhejiang University, Lishui, 323000, China; Department of Radiology, The Fifth Affiliated Hospital of Wenzhou Medical University, Lishui, 323000, China
| | - Min Xu
- Key Laboratory of Imaging Diagnosis and Minimally Invasive Intervention Research, Lishui Hospital, School of Medicine, Zhejiang University, Lishui, 323000, China; Department of Radiology, The Fifth Affiliated Hospital of Wenzhou Medical University, Lishui, 323000, China
| | - Zhongwei Zhao
- Key Laboratory of Imaging Diagnosis and Minimally Invasive Intervention Research, Lishui Hospital, School of Medicine, Zhejiang University, Lishui, 323000, China; Department of Radiology, The Fifth Affiliated Hospital of Wenzhou Medical University, Lishui, 323000, China.
| | - Jiansong Ji
- Key Laboratory of Imaging Diagnosis and Minimally Invasive Intervention Research, Lishui Hospital, School of Medicine, Zhejiang University, Lishui, 323000, China; Department of Radiology, The Fifth Affiliated Hospital of Wenzhou Medical University, Lishui, 323000, China.
| |
Collapse
|