1
|
Vairappan B, Mukherjee V, Subramanian SB, Ram AK, Ravikumar TS. Nimbolide attenuates hepatocellular carcinoma by regulating miRNAs 21, 145 and 221 and their target gene expression. Gene 2025; 937:149126. [PMID: 39645097 DOI: 10.1016/j.gene.2024.149126] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2024] [Revised: 11/13/2024] [Accepted: 11/25/2024] [Indexed: 12/09/2024]
Abstract
BACKGROUND AND AIMS MicroRNAs (miRNAs) are becoming progressively emerging in cancer research from an etiologic and curative point of view. Several miRNAs act as oncogenes or tumor suppressors, which are dysregulated in numerous cancers. Our previous studies have established that nimbolide (a bioactive terpenoid from neem) attenuated hepatocellular carcinoma (HCC) through various mechanisms in mice. Here, we aimed to elucidate the effect of nimbolide in modulating specific miRNAs (21, 145, and 221) and their target genes involved in promoting inflammation and cancer cell proliferation in HCC mice. METHODS Following the induction of HCC in mice at 28 weeks, nimbolide (6 mg/kg b.wt.) was administered orally for four consecutive weeks. RESULTS We found significantly increased hepatic expression of miR-21a-3p, miR-21a-5p, miR-221-5p and miR-221-3p whilst significantly decreased miR-145a-5p in HCC mice. Nimbolide treatment to HCC mice substantially reduced the miR-21a-5p and miR-221-3p and improved miR-145a-5p gene expression. Our in-silico study also supports these findings. Moreover, hepatic tight junction (TJ) associated proteins such as claudins 1&5 mRNA and protein were increased considerably, whilst significantly decreased hepatic claudin 2 mRNA and protein expression noted in HCC mice. Nimbolide also regulates cadherins, ROCK 1, MMP 9, cyclin D1, CDK4, NF κB and TNFα mRNA expression in HCC mice. CONCLUSION We identified for the first time that nibmolide treatment to HCC mice significantly attenuated hepatic miRNAs 21 & 221 expressions and sheltered miR-145 expression. These findings were further confirmed with in-silico studies. Moreover, nibmolide treatment in HCC mice regulates miRNA target genes involved in cancer cell proliferation and inflammation, thereby attenuating HCC progression in mice.
Collapse
Affiliation(s)
- Balasubramaniyan Vairappan
- Liver Diseases Research Lab, Department of Biochemistry, Jawaharlal Institute of Postgraduate Medical Education and Research, Dhanvantari Nagar, Puducherry 605006, India.
| | - Victor Mukherjee
- Liver Diseases Research Lab, Department of Biochemistry, Jawaharlal Institute of Postgraduate Medical Education and Research, Dhanvantari Nagar, Puducherry 605006, India
| | - Siva Bala Subramanian
- Liver Diseases Research Lab, Department of Biochemistry, Jawaharlal Institute of Postgraduate Medical Education and Research, Dhanvantari Nagar, Puducherry 605006, India
| | - Amit Kumar Ram
- Liver Diseases Research Lab, Department of Biochemistry, Jawaharlal Institute of Postgraduate Medical Education and Research, Dhanvantari Nagar, Puducherry 605006, India
| | - T S Ravikumar
- All India Institute of Medical Sciences (AIIMS), Mangalagiri, Andhra Pradesh, India
| |
Collapse
|
2
|
Jogi M, Asnani H, Singh S, Kumar P. Nimbolide: A Potential Phytochemical Agent in Multimodal Pancreatic Cancer Therapies. Mini Rev Med Chem 2025; 25:27-41. [PMID: 38874049 DOI: 10.2174/0113895575293138240527061556] [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: 12/19/2023] [Revised: 04/09/2024] [Accepted: 04/20/2024] [Indexed: 06/15/2024]
Abstract
A significant contributor to cancer-related death, pancreatic cancer (PC) has a terrible prognosis in general that has not altered over many years. Currently, it is extremely difficult to prevent disease or discover it early enough to initiate treatment. PC is a challenging malignancy to treat, and several major impediments significantly impact the effectiveness of its treatment. These obstacles primarily include chemoresistance, drug toxicity, and limited drug bioavailability. Phytochemicals can be used as an alternative to chemotherapeutic drugs, or they can augment the anticancer properties of the chemotherapeutic agents. Nimbolide (NL) is a prominent limonoid compound found in Azadirachta indica, and has garnered substantial attention as a phytochemical with anticancer potential. It has powerful antiproliferative effects on a variety of cancer cell lines and is effective as a chemotherapeutic in preclinical studies. The primary modes of action of NL include suppression of metastasis and angiogenesis, activation of apoptosis, anti-proliferation, and control of enzymes that metabolize carcinogens. Despite numerous pharmacodynamic (PD) investigations, NL is still in the early stages of the drug development process because no comprehensive pharmacokinetic studies or long-term toxicity studies. Preclinical and toxicological assessments should be conducted to establish an appropriate dosage range, ensuring the safety of NL for its application in initial human clinical trials. This review endeavors to provide a comprehensive summary of the current developmental stage of NL along with nanoparticles as a principal candidate for therapeutic purposes in PC.
Collapse
Affiliation(s)
- Mukesh Jogi
- Division of Molecular Biology, ICMR-National Institute of Cancer Prevention and Research (NICPR), NOIDA, India
- Amity Institute of Biotechnology, Amity University, NOIDA, India
- Department of Biotecnology ICMR- National Institute for Research in Environmental Health, Bhopal, India
| | - Hitakshi Asnani
- Banasthli Vidyapith, Radha Kishnpura, Rajasthan, 304022, India
| | - Sohini Singh
- Amity Institute of Biotechnology, Amity University, NOIDA, India
| | - Pramod Kumar
- Division of Molecular Biology, ICMR-National Institute of Cancer Prevention and Research (NICPR), NOIDA, India
| |
Collapse
|
3
|
Arvindh S, Priyadarshini M, Baba AB, Veeravarmal V, Mishra R, Dash R, Nagini S. The Neem Limonoid Nimbolide Modulates Key Components of the DNA Damage Response Signalling in Cellular and Animal Models of Oral Squamous Cell Carcinoma. Curr Pharm Biotechnol 2025; 26:428-442. [PMID: 38561609 DOI: 10.2174/0113892010291998240321074920] [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: 11/14/2023] [Revised: 02/02/2024] [Accepted: 02/12/2024] [Indexed: 04/04/2024]
Abstract
BACKGROUND Deregulated DNA damage response (DDR) network is implicated in cancer progression and therapy resistance. OBJECTIVE The present study was designed to investigate whether nimbolide, an anticancer neem limonoid, targets key components of the DDR signalling pathway in cellular and animal models of oral squamous cell carcinoma (OSCC). METHODS OSCC cells (SCC-4 and SCC-9), 7,12-dimethylbenz[a]anthracene (DMBA)-induced hamster buccal pouch (HBP) carcinoma model, chemoresistant OSCC patient-derived xenograft (PDX) model established in athymic nude mice, and tissue sections from patients with oral premalignant/malignant disease were used for the study. Key molecules that orchestrate the DDR, including the MRN complex, ATM, DNA-PKcs, H2AX, and p53, were analysed by qRTPCR, immunoblotting, immunofluorescence, and immunohistochemistry. Cell proliferation and apoptosis indices were evaluated. RESULTS Nimbolide significantly reduced 8-oxodG levels, expression of MRN, ATMS1891, and γ- H2AX, with an increase in p-p53S15 in OSCC cells as well as in the HBP model. Nimbolide potentiated the effect of KU-55933 in ATM inhibition. In the PDX model, nimbolide suppressed tumor formation, stimulated DDR and apoptosis, inhibited cell proliferation, and enhanced sensitivity to cisplatin. Analysis of p-ATM expression revealed a significant increase during the sequential progression of hamster and human OSCC. CONCLUSION This study provides compelling evidence that nimbolide functions as a DDR inhibitor in cellular and hamster OSCC models and as a DDR activator in the PDX model primarily by targeting ATM. Small molecules like nimbolide that modulate DDR are of immense benefit in cancer therapy. The study has also unveiled p-ATM as a promising biomarker of tumour progression in human OSCCs.
Collapse
Affiliation(s)
- Soundararajan Arvindh
- Department of Biochemistry & Biotechnology, Faculty of Science, Annamalai University, Annamalainagar, 608002, Tamil Nadu, India
| | | | - Abdul Basit Baba
- Advanced Centre for Human Genetics, Sher-i-Kashmir Institute of Medical Sciences (SKIMS), Srinagar, Jammu & Kashmir, 190011, India
| | - Veeran Veeravarmal
- Division of Oral and Maxillofacial Pathology, Government Dental College and Hospital, Cuddalore District, 608 002, Tamil Nadu, India
| | - Rajakishore Mishra
- Department of Life Sciences, School of Natural Sciences, Central University of Jharkhand, Ratu-Lohardaga Road, Brambe, Ranchi, 835205, Jharkhand, India
| | - Rupesh Dash
- Institute of Life Sciences, Bhubaneswar, Odisha, 751023, India
| | - Siddavaram Nagini
- Department of Biochemistry & Biotechnology, Faculty of Science, Annamalai University, Annamalainagar, 608002, Tamil Nadu, India
| |
Collapse
|
4
|
Markowitsch SD, Pham T, Rutz J, Chun FKH, Haferkamp A, Tsaur I, Juengel E, Ries N, Thomas A, Blaheta RA. Growth of Renal Cancer Cell Lines Is Strongly Inhibited by Synergistic Activity of Low-Dosed Amygdalin and Sulforaphane. Nutrients 2024; 16:3750. [PMID: 39519581 PMCID: PMC11547972 DOI: 10.3390/nu16213750] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2024] [Revised: 10/29/2024] [Accepted: 10/31/2024] [Indexed: 11/16/2024] Open
Abstract
Background: Plant derived isolated compounds or extracts enjoy great popularity among cancer patients, although knowledge about their mode of action is unclear. The present study investigated whether the combination of two herbal drugs, the cyanogenic diglucoside amygdalin and the isothiocyanate sulforaphane (SFN), influences growth and proliferation of renal cell carcinoma (RCC) cell lines. Methods: A498, Caki-1, and KTCTL-26 cells were exposed to low-dosed amygdalin (1 or 5 mg/mL), or SFN (5 µM) or to combined SFN-amygdalin. Tumor growth and proliferation were analyzed by MTT, BrdU incorporation, and clone formation assays. Cell cycle phases and cell cycle-regulating proteins were analyzed by flow cytometry and Western blotting, respectively. The effectiveness of the amygdalin-SFN combination was determined using the Bliss independence model. Results: 1 mg/mL amygdalin or 5 µM SFN, given separately, did not suppress RCC cell growth, and 5 mg/mL amygdalin only slightly diminished A498 (but not Caki-1 and KTCTL-26) cell growth. However, already 1 mg/mL amygdalin potently inhibited growth of all tumor cell lines when combined with SFN. Accordingly, 1 mg/mL amygdalin suppressed BrdU incorporation only when given together with SFN. Clonogenic growth was also drastically reduced by the drug combination, whereas only minor effects were seen under single drug treatment. Superior efficacy of co-treatment, compared to monodrug exposure, was also seen for cell cycling, with an enhanced G0/G1 and diminished G2/M phase in A498 cells. Cell cycle regulating proteins were altered differently, depending on the applied drug schedule (single versus dual application) and the RCC cell line, excepting phosphorylated Akt which was considerably diminished in all three cell lines with maximum effects induced by the drug combination. The Bliss independence analysis verified synergistic interactions between amygdalin and SFN. Conclusions: These results point to synergistic effects of amygdalin and SFN on RCC cell growth and clone formation and Akt might be a relevant target protein. The combined use of low-dosed amygdalin and SFN could, therefore, be beneficial as a complementary option to treat RCC. To evaluate clinical feasibility, the in vitro protocol must be applied to an in vivo model.
Collapse
Affiliation(s)
- Sascha D. Markowitsch
- Department of Urology and Pediatric Urology, University Medical Center Mainz, 55131 Mainz, Germany; (S.D.M.); (J.R.); (A.H.); (I.T.); (E.J.); (N.R.); (A.T.)
| | - Thao Pham
- Department of Urology, Goethe-University, 60590 Frankfurt am Main, Germany; (T.P.); (F.K.-H.C.)
| | - Jochen Rutz
- Department of Urology and Pediatric Urology, University Medical Center Mainz, 55131 Mainz, Germany; (S.D.M.); (J.R.); (A.H.); (I.T.); (E.J.); (N.R.); (A.T.)
| | - Felix K.-H. Chun
- Department of Urology, Goethe-University, 60590 Frankfurt am Main, Germany; (T.P.); (F.K.-H.C.)
| | - Axel Haferkamp
- Department of Urology and Pediatric Urology, University Medical Center Mainz, 55131 Mainz, Germany; (S.D.M.); (J.R.); (A.H.); (I.T.); (E.J.); (N.R.); (A.T.)
| | - Igor Tsaur
- Department of Urology and Pediatric Urology, University Medical Center Mainz, 55131 Mainz, Germany; (S.D.M.); (J.R.); (A.H.); (I.T.); (E.J.); (N.R.); (A.T.)
| | - Eva Juengel
- Department of Urology and Pediatric Urology, University Medical Center Mainz, 55131 Mainz, Germany; (S.D.M.); (J.R.); (A.H.); (I.T.); (E.J.); (N.R.); (A.T.)
| | - Nathalie Ries
- Department of Urology and Pediatric Urology, University Medical Center Mainz, 55131 Mainz, Germany; (S.D.M.); (J.R.); (A.H.); (I.T.); (E.J.); (N.R.); (A.T.)
| | - Anita Thomas
- Department of Urology and Pediatric Urology, University Medical Center Mainz, 55131 Mainz, Germany; (S.D.M.); (J.R.); (A.H.); (I.T.); (E.J.); (N.R.); (A.T.)
| | - Roman A. Blaheta
- Department of Urology and Pediatric Urology, University Medical Center Mainz, 55131 Mainz, Germany; (S.D.M.); (J.R.); (A.H.); (I.T.); (E.J.); (N.R.); (A.T.)
| |
Collapse
|
5
|
Yang S, Li D. Role of microRNAs in triple‑negative breast cancer and new therapeutic concepts (Review). Oncol Lett 2024; 28:431. [PMID: 39049985 PMCID: PMC11268089 DOI: 10.3892/ol.2024.14565] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2024] [Accepted: 06/27/2024] [Indexed: 07/27/2024] Open
Abstract
Breast cancer has surpassed lung cancer as the most prevalent malignancy affecting women worldwide. Triple-negative breast cancer (TNBC) is the type of breast cancer with the worst prognosis. As a heterogeneous disease, TNBC has a pathogenesis that involves multiple oncogenic pathways, including involvement of gene mutations and alterations in signaling pathways. MicroRNAs (miRNAs) are small endogenous, single-stranded non-coding RNAs that bind to the 3' untranslated region of target cell mRNAs to negatively regulate the gene expression of these specific mRNAs. Therefore, miRNAs are involved in cell growth, development, division and differentiation stages. miRNAs are also involved in gene targeting in tumorigenesis, tumor growth and the regulation of metastasis, including in breast cancer. Meanwhile, miRNAs also regulate components of signaling pathways. In this review, the role of miRNAs in the TNBC signaling pathway discovered in recent years is described in detail. The new concept of bi-targeted therapy for breast cancer using miRNA and artificial intelligence is also discussed.
Collapse
Affiliation(s)
- Shaofeng Yang
- Department of Thyroid and Breast Surgery, Affiliated Hospital of Inner Mongolia Medical University, Inner Mongolia Medical University, Hohhot, Inner Mongolia Autonomous Region 010050, P.R. China
| | - Donghai Li
- Department of Thyroid and Breast Surgery, Affiliated Hospital of Inner Mongolia Medical University, Inner Mongolia Medical University, Hohhot, Inner Mongolia Autonomous Region 010050, P.R. China
| |
Collapse
|
6
|
Rajendran P, Renu K, Abdallah BM, Ali EM, Veeraraghavan VP, Sivalingam K, Rustagi Y, Abdelsalam SA, Ibrahim RIH, Al-Ramadan SY. Nimbolide: promising agent for prevention and treatment of chronic diseases (recent update). Food Nutr Res 2024; 68:9650. [PMID: 38571915 PMCID: PMC10989234 DOI: 10.29219/fnr.v68.9650] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2023] [Revised: 10/09/2023] [Accepted: 10/16/2023] [Indexed: 04/05/2024] Open
Abstract
Background Nimbolide, a bioactive compound derived from the neem tree, has garnered attention as a potential breakthrough in the prevention and treatment of chronic diseases. Recent updates in research highlight its multifaceted pharmacological properties, demonstrating anti-inflammatory, antioxidant, and anticancer effects. With a rich history in traditional medicine, nimbolide efficacy in addressing the molecular complexities of conditions such as cardiovascular diseases, diabetes, and cancer positions it as a promising candidate for further exploration. As studies progress, the recent update underscores the growing optimism surrounding nimbolide as a valuable tool in the ongoing pursuit of innovative therapeutic strategies for chronic diseases. Methods The comprehensive search of the literature was done until September 2020 on the MEDLINE, Embase, Scopus and Web of Knowledge databases. Results Most studies have shown the Nimbolide is one of the most potent limonoids derived from the flowers and leaves of neem (Azadirachta indica), which is widely used to treat a variety of human diseases. In chronic diseases, nimbolide reported to modulate the key signaling pathways, such as Mitogen-activated protein kinases (MAPKs), Wingless-related integration site-β (Wnt-β)/catenin, NF-κB, PI3K/AKT, and signaling molecules, such as transforming growth factor (TGF-β), Matrix metalloproteinases (MMPs), Vascular Endothelial Growth Factor (VEGF), inflammatory cytokines, and epithelial-mesenchymal transition (EMT) proteins. Nimbolide has anti-inflammatory, anti-microbial, and anti-cancer properties, which make it an intriguing compound for research. Nimbolide demonstrated therapeutic potential for osteoarthritis, rheumatoid arthritis, cardiovascular, inflammation and cancer. Conclusion The current review mainly focused on understanding the molecular mechanisms underlying the therapecutic effects of nimbolide in chronic diseases.
Collapse
Affiliation(s)
- Peramaiyan Rajendran
- Department of Biological Sciences, College of Science, King Faisal University, Al Ahsa, Saudi Arabia
- Centre of Molecular Medicine and Diagnostics (COMManD), Department of Biochemistry, Saveetha Dental College & Hospitals, Saveetha Institute of Medical and Technical Sciences, Saveetha University, Chennai, Tamil Nadu, India
| | - Kaviyarasi Renu
- Centre of Molecular Medicine and Diagnostics (COMManD), Department of Biochemistry, Saveetha Dental College & Hospitals, Saveetha Institute of Medical and Technical Sciences, Saveetha University, Chennai, Tamil Nadu, India
| | - Basem M. Abdallah
- Department of Biological Sciences, College of Science, King Faisal University, Al Ahsa, Saudi Arabia
| | - Enas M. Ali
- Department of Biological Sciences, College of Science, King Faisal University, Al Ahsa, Saudi Arabia
- Department of Botany and Microbiology, Faculty of Science, Cairo University, Cairo, Egypt
| | - Vishnu Priya Veeraraghavan
- Centre of Molecular Medicine and Diagnostics (COMManD), Department of Biochemistry, Saveetha Dental College & Hospitals, Saveetha Institute of Medical and Technical Sciences, Saveetha University, Chennai, Tamil Nadu, India
| | - Kalaiselvi Sivalingam
- Department of Developmental, Molecular and Chemical Biology, Tufts University School of Medicine, Boston, MA, USA
| | - Yashika Rustagi
- Centre for Cancer Genomics, Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA, USA
| | - Salaheldin Abdelraouf Abdelsalam
- Department of Biological Sciences, College of Science, King Faisal University, Al Ahsa, Saudi Arabia
- Department of Zoology, Faculty of Science, Assiut University, Assiut, Egypt
| | - Rashid Ismael Hag Ibrahim
- Department of Biological Sciences, College of Science, King Faisal University, Al Ahsa, Saudi Arabia
- Department of Botany, Faculty of Science, University of Khartoum, Sudan
| | - Saeed Yaseen Al-Ramadan
- Department of Anatomy, College of Veterinary Medicine, King Faisal University, Al-Ahsa, Saudi Arabia
| |
Collapse
|
7
|
Nagini S, Palrasu M, Bishayee A. Limonoids from neem (Azadirachta indica A. Juss.) are potential anticancer drug candidates. Med Res Rev 2024; 44:457-496. [PMID: 37589457 DOI: 10.1002/med.21988] [Citation(s) in RCA: 9] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2022] [Revised: 07/06/2023] [Accepted: 08/06/2023] [Indexed: 08/18/2023]
Abstract
Neem (Azadirachta indica A. Juss.), a versatile evergreen tree recognized for its ethnopharmacological value, is a rich source of limonoids of the triterpenoid class, endowed with potent medicinal properties. Extracts of neem have been documented to display anticancer effects in diverse malignant cell lines as well as in preclinical animal models that has largely been attributed to the constituent limonoids. Of late, neem limonoids have become the cynosure of research attention as potential candidate agents for cancer prevention and therapy. Among the various limonoids found in neem, azadirachtin, epoxyazadiradione, gedunin, and nimbolide, have been extensively investigated for anticancer activity. Azadirachtin, a potent biodegradable pesticide, exhibits profound antiproliferative effects by preventing mitotic spindle formation and cell division. The antiproliferative activity of gedunin has been demonstrated to be mediated primarily via inhibition of heat shock protein90 and its client proteins. Epoxyazadiradione inhibits pro-inflammatory and kinase-driven signaling pathways to block tumorigenesis. Nimbolide, the most potent cytotoxic neem limonoid, inhibits the growth of cancer cells by regulating the phosphorylation of keystone kinases that drive oncogenic signaling besides modulating the epigenome. There is overwhelming evidence to indicate that neem limonoids exert anticancer effects by preventing the acquisition of hallmark traits of cancer, such as cell proliferation, apoptosis evasion, inflammation, invasion, angiogenesis, and drug resistance. Neem limonoids are value additions to the armamentarium of natural compounds that target aberrant oncogenic signaling to inhibit cancer development and progression.
Collapse
Affiliation(s)
- Siddavaram Nagini
- Department of Biochemistry & Biotechnology, Faculty of Science, Annamalai University, Annamalainagar, Tamil Nadu, India
| | - Manikandan Palrasu
- Department of Pathology, Microbiology, and Immunology, University of South Carolina School of Medicine, Columbia, South Carolina, USA
| | - Anupam Bishayee
- College of Osteopathic Medicine, Lake Erie College of Osteopathic Medicine, Bradenton, Florida, USA
| |
Collapse
|
8
|
Cao D, Zhou L, Hu R. Exosomes derived from BMSCs alleviates high glucose-induced diabetic retinopathy via carrying miR-483-5p. J Biochem Mol Toxicol 2024; 38:e23616. [PMID: 38069837 DOI: 10.1002/jbt.23616] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2022] [Revised: 07/26/2023] [Accepted: 11/28/2023] [Indexed: 01/18/2024]
Abstract
Diabetic retinopathy (DR) is a progressive disease which can cause health problem. It has been reported that bone marrow mesenchymal stem cells (BMSCs)-secreted exosomes could regulate the progression of DR via carrying microRNAs. Meanwhile, miR-483-5p was downregulated in DR; however, whether BMSCs-secreted exosomes can modulate DR progression via carrying miR-483-5p remains unclear. To mimic DR in vitro, ARPE-19 cells were exposed to 30 mM high glucose (HG). Exosomes were isolated from BMSCs and identified by transmission electron microscopy, nanoparticle tracking analysis, and western blot. Cell counting kit-8 assay was applied for assessing the cell viability. Flow cytometry was applied to test the cell apoptosis. Meanwhile, dual luciferase assay was used to evaluate the association among miR-483-5p and downstream target insulin-like growth factor 1 receptor (IGF-1R). In addition, quantitative reverse-transcription polymerase chain reaction and western blot were used for exploring the level of miR-483-5p and IGF-1R. HG significantly induced apoptosis in ARPE-19 cells, while BMSCs-derived exosomes reversed this phenomenon. In addition, inhibition of miR-483-5p expression of exosomes further aggravated HG-induced ARPE-19 cell apoptosis. Meanwhile, IGF-1R was the downstream messenger RNA of miR-483-5p, and IGF-1R silencing could reverse the effect of exosomes with downregulated miR-483-5p on HG-induced cell injury. Exosomes derived from BMSCs inhibit the progression of DR via carrying miR-483-5p. Thus, our study might provide a theoretical basis for discovering new strategies against DR.
Collapse
Affiliation(s)
- Dan Cao
- Department of Ophthalmology, The Second Xiangya Hospital of Central South University, Changsha, Hunan Province, People's Republic of China
| | - Liang Zhou
- Department of Ophthalmology, The Second Xiangya Hospital of Central South University, Changsha, Hunan Province, People's Republic of China
| | - Rong Hu
- Department of Ophthalmology, Hunan Provincial People's Hospital, The First Affiliated hospital of Hunan Normal University, Changsha, Hunan Province, People's Republic of China
| |
Collapse
|
9
|
Mohapatra P, Madhulika S, Behera S, Singh P, Sa P, Prasad P, Swain RK, Sahoo SK. Nimbolide-based nanomedicine inhibits breast cancer stem-like cells by epigenetic reprogramming of DNMTs-SFRP1-Wnt/β-catenin signaling axis. MOLECULAR THERAPY. NUCLEIC ACIDS 2023; 34:102031. [PMID: 37771911 PMCID: PMC10523002 DOI: 10.1016/j.omtn.2023.102031] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 02/11/2023] [Accepted: 09/06/2023] [Indexed: 09/30/2023]
Abstract
Triple-negative breast cancer (TNBC) harbors a high percentage of breast cancer stem-like cells (BCSCs) that significantly contribute to poor prognosis, metastasis, and relapse of the disease. Thus, targeting BCSCs could be a promising approach to combat TNBC. In this context, we investigated nimbolide (Nim), a limonoid triterpenoid that has potent anticancer properties, but poor pharmacokinetics and low bioavailability limit its therapeutic application. So, to enhance the therapeutic potential of Nim, Nim-encapsulated poly(lactic-co-glycolic acid) (PLGA) nanoparticles (Nim NPs) were formulated and the anticancer stem cell (CSC) effects evaluated in vitro and in vivo. In vitro studies suggested that Nim NPs significantly inhibited several inherent characteristics of BCSCs, such as stemness, self-renewability, chemoresistance, epithelial-to-mesenchymal transition (EMT), and migration in comparison to native Nim. Next, the mechanism behind the anti-CSC effect of Nim was explored. Mechanistically, we found that Nim epigenetically restores tumor suppressor gene secreted frizzled-related protein 1 (SFRP1) expression by downregulating DNA methyltransferases (DNMTs), leading to Wnt/β-catenin signaling inhibition. Further, in vivo results demonstrated that Nim NPs showed enhanced anti-tumor and anti-metastatic effects compared to native Nim in two preclinical models without any systemic toxicity. Overall, these findings provide proof of concept that Nim-based phytonanomedicine can inhibit BCSCs by epigenetic reprogramming of the DNMTs-SFRP1-Wnt/β-catenin signaling axis.
Collapse
Affiliation(s)
- Priyanka Mohapatra
- Institute of Life Sciences, Nalco Square, Bhubaneswar 751023, Odisha, India
- Regional Centre for Biotechnology, Faridabad 121001, Haryana, India
| | - Swati Madhulika
- Institute of Life Sciences, Nalco Square, Bhubaneswar 751023, Odisha, India
- Regional Centre for Biotechnology, Faridabad 121001, Haryana, India
| | - Somalisa Behera
- Institute of Life Sciences, Nalco Square, Bhubaneswar 751023, Odisha, India
| | - Priya Singh
- Institute of Life Sciences, Nalco Square, Bhubaneswar 751023, Odisha, India
- Regional Centre for Biotechnology, Faridabad 121001, Haryana, India
| | - Pratikshya Sa
- Institute of Life Sciences, Nalco Square, Bhubaneswar 751023, Odisha, India
- Regional Centre for Biotechnology, Faridabad 121001, Haryana, India
| | - Punit Prasad
- Institute of Life Sciences, Nalco Square, Bhubaneswar 751023, Odisha, India
| | - Rajeeb Kumar Swain
- Institute of Life Sciences, Nalco Square, Bhubaneswar 751023, Odisha, India
| | | |
Collapse
|
10
|
Li X, Mei M, Pu X, Chen X, Li X, Meng F, He S, Li J, Gu W, Yang X, Zhang F, Yu J. Protective effect and mechanism of Polygonatum kingianum against hypoxia-induced injury. Heliyon 2023; 9:e14353. [PMID: 36967867 PMCID: PMC10034467 DOI: 10.1016/j.heliyon.2023.e14353] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2022] [Revised: 02/24/2023] [Accepted: 03/02/2023] [Indexed: 03/11/2023] Open
Abstract
Background Hypoxia is an essential cause of fatigue and aging, and is associated with the occurrence and development of many diseases. Polygonatum kingianum (PK) is a deficiency-nourishing Chinese herbal medicine utilized as both medicine and food, and it has long been used to ameliorate human conditions associated with fatigue and aging over 2000 years in China. PK is an important genuine-medicinal-materials cultivated in Yunnan, China, and is used by the Bai, Wa, and Zhuang nationalities as a traditional medicine for enhancing immunity, anti-fatigue, and anti-aging, while the preventive effect of PK on hypoxia-induced injury and the underlying mechanism are indefinite. Aim of the study The present study aimed to evaluate the anti-hypoxia efficacy and understand the corresponding mechanism of PK water extract. Materials and methods The main active ingredients and targets of PK were predicted using network pharmacology, and the anti-hypoxia activities of Gracillin and Liquiritigenin were verified by in vitro experiments. The pharmacodynamic experiments were conducted to evaluate the major signal pathways of PK for detecting anti-hypoxia activity. Results Fifty active ingredients and 371 potential targets were screened by network pharmacology, then, we confirmed that Gracillin and Liquiritigenin were the main active components of PK to exert anti-hypoxia effect in vitro. The pharmacodynamic experiments revealed that PK enhanced the extension rate of the survival time (ERST) and regulated the targets-related biochemical parameters of rats under hypoxia, showing significant anti-hypoxia effects on rats. Conclusion The network pharmacology results suggested that PK exerts its anti-hypoxia effect through a multi-component and multi-target manner. Simultaneously, we also observed that Gracillin (saponins) and Liquiritigenin (flavonoids) are the main active components of PK to play a role in anti-hypoxia. The anti-hypoxia effect of PK could be associated with scavenging excess free radicals, maintaining the activities of antioxidant enzymes, and inhibiting oxidative stress due to lipid peroxidation. These findings provide insight into the Polygonatum kingianum as promising medicines or healthcare products for preventing and treating hypoxia.
Collapse
|