1
|
Wang C, Feng Q, Shi S, Qin Y, Lu H, Zhang P, Liu J, Chen B. The Rational Engineered Bacteria Based Biohybrid Living System for Tumor Therapy. Adv Healthc Mater 2024; 13:e2401538. [PMID: 39051784 DOI: 10.1002/adhm.202401538] [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: 04/26/2024] [Revised: 07/16/2024] [Indexed: 07/27/2024]
Abstract
Living therapy based on bacterial cells has gained increasing attention for their applications in tumor treatments. Bacterial cells can naturally target to tumor sites and active the innate immunological responses. The intrinsic advantages of bacteria attribute to the development of biohybrid living carriers for targeting delivery toward hypoxic environments. The rationally engineered bacterial cells integrate various functions to enhance the tumor therapy and reduce toxic side effects. In this review, the antitumor effects of bacteria and their application are discussed as living therapeutic agents across multiple antitumor platforms. The various kinds of bacteria used for cancer therapy are first introduced and demonstrated the mechanism of antitumor effects as well as the immunological effects. Additionally, this study focused on the genetically modified bacteria for the production of antitumor agents as living delivery system to treat cancer. The combination of living bacterial cells with functional nanomaterials is then discussed in the cancer treatments. In brief, the rational design of living therapy based on bacterial cells highlighted a rapid development in tumor therapy and pointed out the potentials in clinical applications.
Collapse
Affiliation(s)
- Chen Wang
- School of Biomedical Engineering, Shenzhen Campus of Sun Yat-Sen University, No. 66, Gongchang Road, Guangming District, Shenzhen, Guangdong, 518107, China
| | - Qiliner Feng
- School of Biomedical Engineering, Shenzhen Campus of Sun Yat-Sen University, No. 66, Gongchang Road, Guangming District, Shenzhen, Guangdong, 518107, China
| | - Si Shi
- School of Biomedical Engineering, Shenzhen Campus of Sun Yat-Sen University, No. 66, Gongchang Road, Guangming District, Shenzhen, Guangdong, 518107, China
| | - Yuxuan Qin
- School of Biomedical Engineering, Shenzhen Campus of Sun Yat-Sen University, No. 66, Gongchang Road, Guangming District, Shenzhen, Guangdong, 518107, China
| | - Hongli Lu
- School of Biomedical Engineering, Shenzhen Campus of Sun Yat-Sen University, No. 66, Gongchang Road, Guangming District, Shenzhen, Guangdong, 518107, China
| | - Peng Zhang
- School of Biomedical Engineering, Shenzhen Campus of Sun Yat-Sen University, No. 66, Gongchang Road, Guangming District, Shenzhen, Guangdong, 518107, China
| | - Jie Liu
- School of Biomedical Engineering, Shenzhen Campus of Sun Yat-Sen University, No. 66, Gongchang Road, Guangming District, Shenzhen, Guangdong, 518107, China
| | - Baizhu Chen
- School of Biomedical Engineering, Shenzhen Campus of Sun Yat-Sen University, No. 66, Gongchang Road, Guangming District, Shenzhen, Guangdong, 518107, China
- Guangdong Provincial Key Laboratory of Sensor Technology and Biomedical Instrument, Sun Yat-Sen University, Shenzhen, Guangdong, 518107, China
| |
Collapse
|
2
|
Kokhdan EP, Khodavandi P, Ataeyan MH, Alizadeh F, Khodavandi A, Zaheri A. Anti-cancer activity of secreted aspartyl proteinase protein from Candida tropicalis on human cervical cancer HeLa cells. Toxicon 2024; 249:108073. [PMID: 39153686 DOI: 10.1016/j.toxicon.2024.108073] [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: 03/06/2024] [Revised: 07/16/2024] [Accepted: 08/14/2024] [Indexed: 08/19/2024]
Abstract
Cervical cancer is the fourth leading cause of cancer-related death in women worldwide. Microbial products are valuable sources of anti-cancer drugs. The aim of this study was to isolate secreted aspartyl proteinase protein from Candida tropicalis, investigate its inhibitory effect on human cervical cancer HeLa cells, and analyze the expression profiling of selected nuclear stem cell-associated transcription factors. The presence of secreted aspartyl proteinase protein was confirmed by the expression of SAP2 and SAP4 genes in C. tropicalis during the yeast-hyphae transition phase. The enzyme was purified and characterized using the aqueous two-phase system purification method, as well as proteolytic activity and the Bradford and micro-Kjeldahl methods, respectively. The in vitro anti-cancer properties of secreted aspartyl proteinase protein were evaluated by MTT assay, microscopic image analysis, nitric oxide (NO) scavenging activity assay, intracellular reactive oxygen species (ROS) production assay, and RT-qPCR. The isolated C. tropicalis secreted aspartyl proteinase protein exhibited proteinase activity with values ranging from 93.72 to 130.70 μg/mL and 89.88-127.72 μg/mL according to the Bradford and micro-Kjeldahl methods, respectively. Secreted aspartyl proteinase showed effective cytotoxicity in HeLa cell line leading to significant morphological changes. Additionally, it exhibited increased free radical scavenging activity compared to the untreated control group, as evidenced by nitrite inhibition. ROS production increased in HeLa cells exposed to secreted aspartyl proteinase. The expression levels of the nuclear stem cell-associated transcription factors octamer-binding transcription factor 4 (OCT4), sex determining region Y-box 2 (SOX2), and Nanog homeobox (NANOG) were significantly downregulated in the HeLa cells treated with secreted aspartyl proteinase. Secreted aspartyl proteinase protein may be a promising anti-cancer agent, as it effectively affects gene expression and may ultimately reduce the development and progression of cervical cancer. Targeting the genes related to nuclear stem cell-associated transcription factors may provide a novel amenable to cancer treatment.
Collapse
Affiliation(s)
| | | | | | - Fahimeh Alizadeh
- Department of Biology, Gachsaran Branch, Islamic Azad University, Gachsaran, Iran.
| | - Alireza Khodavandi
- Department of Biology, Gachsaran Branch, Islamic Azad University, Gachsaran, Iran.
| | - Ahmad Zaheri
- Department of Biology, Payame Noor University, Tehran, Iran
| |
Collapse
|
3
|
Andreani T, Cheng R, Elbadri K, Ferro C, Menezes T, Dos Santos MR, Pereira CM, Santos HA. Natural compounds-based nanomedicines for cancer treatment: Future directions and challenges. Drug Deliv Transl Res 2024; 14:2845-2916. [PMID: 39003425 PMCID: PMC11385056 DOI: 10.1007/s13346-024-01649-z] [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] [Accepted: 06/05/2024] [Indexed: 07/15/2024]
Abstract
Several efforts have been extensively accomplished for the amelioration of the cancer treatments using different types of new drugs and less invasives therapies in comparison with the traditional therapeutic modalities, which are widely associated with numerous drawbacks, such as drug resistance, non-selectivity and high costs, restraining their clinical response. The application of natural compounds for the prevention and treatment of different cancer cells has attracted significant attention from the pharmaceuticals and scientific communities over the past decades. Although the use of nanotechnology in cancer therapy is still in the preliminary stages, the application of nanotherapeutics has demonstrated to decrease the various limitations related to the use of natural compounds, such as physical/chemical instability, poor aqueous solubility, and low bioavailability. Despite the nanotechnology has emerged as a promise to improve the bioavailability of the natural compounds, there are still limited clinical trials performed for their application with various challenges required for the pre-clinical and clinical trials, such as production at an industrial level, assurance of nanotherapeutics long-term stability, physiological barriers and safety and regulatory issues. This review highlights the most recent advances in the nanocarriers for natural compounds secreted from plants, bacteria, fungi, and marine organisms, as well as their role on cell signaling pathways for anticancer treatments. Additionally, the clinical status and the main challenges regarding the natural compounds loaded in nanocarriers for clinical applications were also discussed.
Collapse
Affiliation(s)
- Tatiana Andreani
- Chemistry Research Centre (CIQUP) and Institute of Molecular Sciences (IMS), Department of Chemistry and Biochemistry, Faculty of Sciences of University of Porto, Rua Do Campo Alegre s/n, 4169-007, Porto, Portugal
- GreenUPorto-Sustainable Agrifood Production Research Centre & Inov4Agro, Department of Biology, Faculty of Sciences of University of Porto, Rua Campo Alegre s/n, 4169-007, Porto, Portugal
| | - Ruoyu Cheng
- Drug Research Program, Division of Pharmaceutical Chemistry and Technology, Faculty of Pharmacy, University of Helsinki, FI-00014, Helsinki, Finland
- Department of Biomaterials and Biomedical Technology, The Personalized Medicine Research Institute Groningen (PRECISION), University Medical Center Groningen, University of Groningen, 9713 AV, Groningen, The Netherlands
| | - Khalil Elbadri
- Drug Research Program, Division of Pharmaceutical Chemistry and Technology, Faculty of Pharmacy, University of Helsinki, FI-00014, Helsinki, Finland
| | - Claudio Ferro
- Drug Research Program, Division of Pharmaceutical Chemistry and Technology, Faculty of Pharmacy, University of Helsinki, FI-00014, Helsinki, Finland
- Research Institute for Medicines, iMed.Ulisboa, Faculty of Pharmacy, Universidade de Lisboa, 1649-003, Lisbon, Portugal
| | - Thacilla Menezes
- Chemistry Research Centre (CIQUP) and Institute of Molecular Sciences (IMS), Department of Chemistry and Biochemistry, Faculty of Sciences of University of Porto, Rua Do Campo Alegre s/n, 4169-007, Porto, Portugal
| | - Mayara R Dos Santos
- Chemistry Research Centre (CIQUP) and Institute of Molecular Sciences (IMS), Department of Chemistry and Biochemistry, Faculty of Sciences of University of Porto, Rua Do Campo Alegre s/n, 4169-007, Porto, Portugal
| | - Carlos M Pereira
- Chemistry Research Centre (CIQUP) and Institute of Molecular Sciences (IMS), Department of Chemistry and Biochemistry, Faculty of Sciences of University of Porto, Rua Do Campo Alegre s/n, 4169-007, Porto, Portugal
| | - Hélder A Santos
- Drug Research Program, Division of Pharmaceutical Chemistry and Technology, Faculty of Pharmacy, University of Helsinki, FI-00014, Helsinki, Finland.
- Department of Biomaterials and Biomedical Technology, The Personalized Medicine Research Institute Groningen (PRECISION), University Medical Center Groningen, University of Groningen, 9713 AV, Groningen, The Netherlands.
| |
Collapse
|
4
|
Ilic-Tomic T, Kramar A, Kostic M, Vojnovic S, Milovanovic J, Petkovic M, D’Agostino PM, Gulder TAM, Nikodinovic-Runic J. Functionalization of silk with actinomycins from Streptomyces anulatus BV365 for biomedical applications. Front Bioeng Biotechnol 2024; 12:1466757. [PMID: 39364265 PMCID: PMC11447452 DOI: 10.3389/fbioe.2024.1466757] [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: 07/18/2024] [Accepted: 09/05/2024] [Indexed: 10/05/2024] Open
Abstract
Silk, traditionally acclaimed as the "queen of fiber," has been widely used thanks to its brilliant performance such as gentleness, smoothness and comfortableness. Owing to its mechanical characteristics and biocompatibility silk has a definitive role in biomedical applications, both as fibroin and fabric. In this work, the simultaneous dyeing and functionalization of silk fabric with pigments from Streptomyces anulatus BV365 were investigated. This strain produced high amounts of orange extracellular pigments on mannitol-soy flour agar, identified as actinomycin D, C2 and C3. The application of purified actinomycins in the dyeing of multifiber fabric was assessed. Actinomycins exhibited a high affinity towards protein fibers (silk and wool), but washing durability was maintained only with silk. Acidic condition (pH5) and high temperature (65°C) facilitated the silk dyeing. The morphologies and chemical components of the treated silk fabrics were analyzed using scanning electron microscopy and Fourier transform infrared spectroscopy. The results showed the pigments bind to the silk through interaction with the carbonyl group in silk fibroin rendering the functionalized, yet surface that does not cause skin irritation. The treated silk exhibited a remarkable antibacterial effect, while the biocompatibility test performed with 3D-reconstructed human epidermis model indicated safe biological properties, paving the way for future application of this material in medicine.
Collapse
Affiliation(s)
- Tatjana Ilic-Tomic
- Institute of Molecular Genetics and Genetic Engineering, University of Belgrade, Belgrade, Serbia
| | - Ana Kramar
- Department of Textile Engineering, Faculty of Technology and Metallurgy, University of Belgrade, Belgrade, Serbia
- Novel Materials and Nanotechnology Group, Institute of Agrochemistry and Food Technology (IATA), Spanish Council for Scientific Research (CSIC), Paterna, Spain
| | - Mirjana Kostic
- Department of Textile Engineering, Faculty of Technology and Metallurgy, University of Belgrade, Belgrade, Serbia
| | - Sandra Vojnovic
- Institute of Molecular Genetics and Genetic Engineering, University of Belgrade, Belgrade, Serbia
| | - Jelena Milovanovic
- Institute of Molecular Genetics and Genetic Engineering, University of Belgrade, Belgrade, Serbia
| | - Milos Petkovic
- Faculty of Pharmacy, University of Belgrade, Belgrade, Serbia
| | - Paul M. D’Agostino
- Helmholtz Institute for Pharmaceutical Research Saarland (HIPS), Saarbrücken, Saarland, Germany
- Technical University of Dresden, Dresden, Saxony, Germany
| | - Tobias A. M. Gulder
- Helmholtz Institute for Pharmaceutical Research Saarland (HIPS), Saarbrücken, Saarland, Germany
- Technical University of Dresden, Dresden, Saxony, Germany
| | | |
Collapse
|
5
|
Mohammed SM, Al-Saedi HFS, Mohammed AQ, Amir AA, Radi UK, Sattar R, Ahmad I, Ramadan MF, Alshahrani MY, Balasim HM, Alawadi A. Mechanisms of Bleomycin-induced Lung Fibrosis: A Review of Therapeutic Targets and Approaches. Cell Biochem Biophys 2024; 82:1845-1870. [PMID: 38955925 DOI: 10.1007/s12013-024-01384-9] [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] [Accepted: 06/21/2024] [Indexed: 07/04/2024]
Abstract
Pulmonary toxicity is a serious side effect of some specific anticancer drugs. Bleomycin is a well-known anticancer drug that triggers severe reactions in the lungs. It is an approved drug that may be prescribed for the treatment of testicular cancers, Hodgkin's and non-Hodgkin's lymphomas, ovarian cancer, head and neck cancers, and cervical cancer. A large number of experimental studies and clinical findings show that bleomycin can concentrate in lung tissue, leading to massive oxidative stress, alveolar epithelial cell death, the proliferation of fibroblasts, and finally the infiltration of immune cells. Chronic release of pro-inflammatory and pro-fibrotic molecules by immune cells and fibroblasts leads to pneumonitis and fibrosis. Both fibrosis and pneumonitis are serious concerns for patients who receive bleomycin and may lead to death. Therefore, the management of lung toxicity following cancer therapy with bleomycin is a critical issue. This review explains the cellular and molecular mechanisms of pulmonary injury following treatment with bleomycin. Furthermore, we review therapeutic targets and possible promising strategies for ameliorating bleomycin-induced lung injury.
Collapse
Affiliation(s)
- Shaimaa M Mohammed
- Department of Pharmacy, Al- Mustaqbal University College, 51001, Hilla, Babylon, Iraq
| | | | | | - Ahmed Ali Amir
- Department of Medical Laboratories Technology, Al-Nisour University College, Baghdad, Iraq
| | - Usama Kadem Radi
- College of Pharmacy, National University of Science and Technology, Nasiriyah, Dhi Qar, Iraq
| | - Ruaa Sattar
- Al-Hadi University College, Baghdad, 10011, Iraq
| | - Irfan Ahmad
- Department of Clinical Laboratory Sciences, College of Applied Medical Sciences, King Khalid University, Abha, Saudi Arabia
| | | | - Mohammad Y Alshahrani
- Department of Clinical Laboratory Sciences, College of Applied Medical Sciences, King Khalid University, Abha, Saudi Arabia.
| | - Halah Majeed Balasim
- Department of Medical Laboratory Technologies, Al Rafidain University College, Bagdad, Iraq
| | - Ahmed Alawadi
- College of technical engineering, the Islamic University, Najaf, Iraq
- College of technical engineering, the Islamic University of Al Diwaniyah, Al Diwaniyah, Iraq
- College of technical engineering, the Islamic University of Babylon, Hilla, Iraq
| |
Collapse
|
6
|
Sood A, Jothiswaran V, Singh A, Sharma A. Anticancer peptides as novel immunomodulatory therapeutic candidates for cancer treatment. EXPLORATION OF TARGETED ANTI-TUMOR THERAPY 2024; 5:1074-1099. [PMID: 39351437 PMCID: PMC11438574 DOI: 10.37349/etat.2024.00264] [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: 05/24/2024] [Accepted: 07/27/2024] [Indexed: 10/04/2024] Open
Abstract
Cancer remains a concern after years of research in this field. Conventional therapies such as chemotherapy, radiation, and surgery are available for cancer treatment, but they are characterized by various side effects. There are several immunological challenges that make it difficult for the immune system and conventional therapies to treat cancer. Some of these challenges include heterogeneity, resistance to medicines, and cancer relapse. Even advanced treatments like immune checkpoint inhibitors (ICIs), which revolutionized cancer treatment, have associated toxicity and resistance further necessitate the exploration of alternative therapies. Anticancer peptides (ACPs) offer promising potential as cancer-fighting agents and address challenges such as treatment resistance, tumor heterogeneity, and metastasis. Although these peptides exist as components of the defense system in various plants, animals, fungi, etc., but can also be created synthetically and used as a new treatment measure. These peptides possess properties that make them appealing for cancer therapy, such as apoptosis induction, inhibition of angiogenesis, and cell membrane breakdown with low toxicity. Their capacity to specifically target cancer cells selectively holds promise for enhancing treatment environments as well as improving patients' quality of life. This review provides detailed insights into the different prospects of ACPs, including their characterization, use as immunomodulatory agents in cancer treatment, and their mechanistic details after addressing various immunological challenges in existing cancer treatment strategies. In conclusion, ACPs have promising potential as novel cancer therapeutics due to their target specificity and fewer side effects than conventional therapies.
Collapse
Affiliation(s)
- Apurva Sood
- Department of Molecular Biology and Genetic Engineering, School of Bioengineering and Biosciences, Lovely Professional University, Punjab 144411, India
| | - V.V. Jothiswaran
- Biotechnology and Medical Engineering, National Institute of Technology, Rourkela 769005, India
| | - Amrita Singh
- Biotechnology and Medical Engineering, National Institute of Technology, Rourkela 769005, India
| | - Anuradha Sharma
- Department of Molecular Biology and Genetic Engineering, School of Bioengineering and Biosciences, Lovely Professional University, Punjab 144411, India
| |
Collapse
|
7
|
Garai S, Thomas J, Dey P, Das D. LGBM-ACp: an ensemble model for anticancer peptide prediction and in silico screening with potential drug targets. Mol Divers 2024; 28:1965-1981. [PMID: 36637711 DOI: 10.1007/s11030-023-10602-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2022] [Accepted: 01/06/2023] [Indexed: 01/14/2023]
Abstract
Conventional cancer therapies are highly expensive and have serious complications. An alternative approach now emphasizes on the development of small, biologically active peptides without acute toxicity. Experimental screening to find curative anticancer peptides (ACP) often gives rise to multiple obstacles and is time dependent. Consequently, developing an effective computational technique to identify promising ACP candidates prior to preclinical research is in high demand. This study proposed a machine-learning framework that used the light gradient-boosting machine as a classifier and two compositional and two binary profile features as input. The ensemble model displayed an accuracy, MCC, and AUROC of 97.52%, 0.91, and 0.98, respectively, which outclassed most of the existing sequence-based computational tools. A distinct dataset of non-mutagenic, non-toxic, and non-inhibitory Cytochrome P-450 peptides was used to validate the hybrid model. The most relevant ACP in the alternative dataset was compared with two standard ACPs, beta defensin 2, and cecropin-A. Molecular docking of the predicted peptide revealed that it has a strong binding affinity with twenty-five anticancer drug targets, most notably phosphoenolpyruvate carboxykinase (- 7.2 kcal/mol). Additionally, molecular dynamics simulation and principal component analysis supported the stability of the peptide-receptor complex. Overall, the present findings will take a step forward in rational drug design through rapid identification and screening of therapeutic peptides.
Collapse
Affiliation(s)
- Swarnava Garai
- Department of Bioengineering, NIT Agartala, Tripura, 799046, India
| | - Juanit Thomas
- Department of Bioengineering, NIT Agartala, Tripura, 799046, India
| | - Palash Dey
- Civil Engineering Department, The ICFAI University, Tripura, 799210, India
| | - Deeplina Das
- Department of Bioengineering, NIT Agartala, Tripura, 799046, India.
| |
Collapse
|
8
|
Jamal QMS, Ahmad V. Bacterial metabolomics: current applications for human welfare and future aspects. JOURNAL OF ASIAN NATURAL PRODUCTS RESEARCH 2024:1-24. [PMID: 39078342 DOI: 10.1080/10286020.2024.2385365] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/20/2024] [Revised: 07/22/2024] [Accepted: 07/24/2024] [Indexed: 07/31/2024]
Abstract
An imbalanced microbiome is linked to several diseases, such as cancer, inflammatory bowel disease, obesity, and even neurological disorders. Bacteria and their by-products are used for various industrial and clinical purposes. The metabolites under discussion were chosen based on their biological impacts on host and gut microbiota interactions as established by metabolome research. The separation of bacterial metabolites by using statistics and machine learning analysis creates new opportunities for applications of bacteria and their metabolites in the environmental and medical sciences. Thus, the metabolite production strategies, methodologies, and importance of bacterial metabolites for human well-being are discussed in this review.
Collapse
Affiliation(s)
- Qazi Mohammad Sajid Jamal
- Department of Health Informatics, College of Applied Medical Sciences, Qassim University, Buraydah 51452, Saudi Arabia
| | - Varish Ahmad
- Health Information Technology Department, The Applied College, King Abdulaziz University, Jeddah 21589, Saudi Arabia
| |
Collapse
|
9
|
Karadkhelkar NM, Gupta P, Barasa L, Chilamakuri R, Hlordzi CK, Acharekar N, Agarwal S, Chen ZS, Yoganathan S. Chemical Derivatization Leads to the Discovery Of Novel Analogs of Azotochelin, a Natural Siderophore, as Promising Anticancer Agents. ChemMedChem 2024; 19:e202300715. [PMID: 38598189 DOI: 10.1002/cmdc.202300715] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2023] [Revised: 03/21/2024] [Accepted: 04/08/2024] [Indexed: 04/11/2024]
Abstract
Siderophores are structurally unique medicinal natural products and exhibit considerable therapeutic potential. Herein, we report the design and synthesis of azotochelin, a natural siderophore, and an extensive library of azotochelin analogs and their anticancer properties. We modified the carboxylic acid and the aromatic ring of azotochelin using various chemical motifs. We evaluated the cytotoxicity of the compounds against six different cancer cell lines (KB-3-1, SNB-19, MCF-7, K-562, SW-620, and NCI-H460) and a non-cancerous cell line (HEK-293). Among the twenty compounds tested, the IC50 values of nine compounds (14, 32, 35-40, and 54) were between 0.7 and 2.0 μM against a lung cancer cell line (NCI-H460). Moreover, several compounds showed good cytotoxicity profile (IC50 <10 μM) against the tested cancer cell lines. The flow cytometry analysis showed that compounds 36 and 38 induced apoptosis in NCI-H460 in a dose-dependent manner. The cell cycle analysis indicated that compounds 36 and 38 significantly arrested the cell cycle at the S phase to block cancer cell proliferation in the NCI-H460 cell line. The study has produced various novel azotochelin analogs that are potentially effective anticancer agents and lead compounds for further synthetic and medicinal chemistry exploration.
Collapse
Affiliation(s)
- Nishant M Karadkhelkar
- Department of Pharmaceutical Sciences, College of Pharmacy and Health Sciences, St. John's University, 8000 Utopia Parkway, Queens, NY, 11439 (S.Y.)
- Current affiliation: The Scripps Research Institute, 10550 N Torrey Pines Rd., La Jolla, CA, 92037
| | - Pranav Gupta
- Department of Pharmaceutical Sciences, College of Pharmacy and Health Sciences, St. John's University, 8000 Utopia Parkway, Queens, NY, 11439 (S.Y.)
| | - Leonard Barasa
- Department of Pharmaceutical Sciences, College of Pharmacy and Health Sciences, St. John's University, 8000 Utopia Parkway, Queens, NY, 11439 (S.Y.)
- Current affiliation: Department of Biochemistry and Molecular Biotechnology, University of Massachusetts Medical School, Worcester, MA, 01605
| | - Rameswari Chilamakuri
- Department of Pharmaceutical Sciences, College of Pharmacy and Health Sciences, St. John's University, 8000 Utopia Parkway, Queens, NY, 11439 (S.Y.)
| | - Christopher K Hlordzi
- Department of Pharmaceutical Sciences, College of Pharmacy and Health Sciences, St. John's University, 8000 Utopia Parkway, Queens, NY, 11439 (S.Y.)
| | - Nikita Acharekar
- Department of Pharmaceutical Sciences, College of Pharmacy and Health Sciences, St. John's University, 8000 Utopia Parkway, Queens, NY, 11439 (S.Y.)
| | - Saurabh Agarwal
- Department of Pharmaceutical Sciences, College of Pharmacy and Health Sciences, St. John's University, 8000 Utopia Parkway, Queens, NY, 11439 (S.Y.)
| | - Zhe-Sheng Chen
- Department of Pharmaceutical Sciences, College of Pharmacy and Health Sciences, St. John's University, 8000 Utopia Parkway, Queens, NY, 11439 (S.Y.)
| | - Sabesan Yoganathan
- Department of Pharmaceutical Sciences, College of Pharmacy and Health Sciences, St. John's University, 8000 Utopia Parkway, Queens, NY, 11439 (S.Y.)
| |
Collapse
|
10
|
Marey MA, Abozahra R, El-Nikhely NA, Kamal MF, Abdelhamid SM, El-Kholy MA. Transforming microbial pigment into therapeutic revelation: extraction and characterization of pyocyanin from Pseudomonas aeruginosa and its therapeutic potential as an antibacterial and anticancer agent. Microb Cell Fact 2024; 23:174. [PMID: 38867319 PMCID: PMC11170807 DOI: 10.1186/s12934-024-02438-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2024] [Accepted: 05/23/2024] [Indexed: 06/14/2024] Open
Abstract
BACKGROUND The objectives of the current study were to extract pyocyanin from Pseudomonas aeruginosa clinical isolates, characterize its chemical nature, and assess its biological activity against different bacteria and cancer cells. Due to its diverse bioactive properties, pyocyanin, being one of the virulence factors of P. aeruginosa, holds a promising, safe, and available therapeutic potential. METHODS 30 clinical P. aeruginosa isolates were collected from different sources of infections and identified by routine methods, the VITEK 2 compact system, and 16 S rRNA. The phenazine-modifying genes (phzM, phzS) were identified using polymerase chain reaction (PCR). Pyocyanin chemical characterization included UV-Vis spectrophotometry, Fourier Transform Infra-Red spectroscopy (FTIR), Gas Chromatography-Mass Spectrometry (GC-MS), and Liquid Chromatography-Mass Spectrometry (LC-MS). The biological activity of pyocyanin was explored by determining the MIC values against different clinical bacterial strains and assessing its anticancer activity against A549, MDA-MB-231, and Caco-2 cancer cell lines using cytotoxicity, wound healing and colony forming assays. RESULTS All identified isolates harboured at least one of the phzM or phzS genes. The co-presence of both genes was demonstrated in 13 isolates. The UV-VIS absorbance peaks were maxima at 215, 265, 385, and 520 nm. FTIR could identify the characteristic pyocyanin functional groups, whereas both GC-MS and LC-MS elucidated the chemical formula C11H18N2O2, with a molecular weight 210. The quadri-technical analytical approaches confirmed the chemical nature of the extracted pyocyanin. The extract showed broad-spectrum antibacterial activity, with the greatest activity against Bacillus, Staphylococcus, and Streptococcus species (MICs 31.25-125 µg/mL), followed by E. coli isolates (MICs 250-1000 µg/mL). Regarding the anticancer activity, the pyocyanin extract showed IC50 values against A549, MDA-MB-231, and Caco-2 cancer cell lines of 130, 105, and 187.9 µg/mL, respectively. Furthermore, pyocyanin has markedly suppressed colony formation and migratory abilities in these cells. CONCLUSIONS The extracted pyocyanin has demonstrated to be a potentially effective candidate against various bacterial infections and cancers. Hence, the current findings could contribute to producing this natural compound easily through an affordable method. Nonetheless, future studies are required to investigate pyocyanin's effects in vivo and analyse the results of combining it with other traditional antibiotics or anticancer drugs.
Collapse
Affiliation(s)
- Moustafa A Marey
- Department of Microbiology and Biotechnology, Division of Clinical and Biological Sciences, College of Pharmacy, Arab Academy for Science, Technology and Maritime Transport (AASTMT), Abu Kir Campus, P.O. Box 1029, Alexandria, Egypt
| | - Rania Abozahra
- Microbiology and Immunology Department, Faculty of Pharmacy, Damanhour University, Damanhour, Egypt
| | - Nefertiti A El-Nikhely
- Department of Biotechnology, Institute of Graduate Studies and Research, Alexandria University, Alexandria, Egypt
| | - Miranda F Kamal
- Department of Pharmaceutical Analytical Chemistry, Faculty of Pharmacy, Damanhour University, Beheira, Egypt
| | - Sarah M Abdelhamid
- Microbiology and Immunology Department, Faculty of Pharmacy, Damanhour University, Damanhour, Egypt
| | - Mohammed A El-Kholy
- Department of Microbiology and Biotechnology, Division of Clinical and Biological Sciences, College of Pharmacy, Arab Academy for Science, Technology and Maritime Transport (AASTMT), Abu Kir Campus, P.O. Box 1029, Alexandria, Egypt.
| |
Collapse
|
11
|
Kerdkumthong K, Chanket W, Runsaeng P, Nanarong S, Songsurin K, Tantimetta P, Angsuthanasombat C, Aroonkesorn A, Obchoei S. Two Recombinant Bacteriocins, Rhamnosin and Lysostaphin, Show Synergistic Anticancer Activity Against Gemcitabine-Resistant Cholangiocarcinoma Cell Lines. Probiotics Antimicrob Proteins 2024; 16:713-725. [PMID: 37294416 DOI: 10.1007/s12602-023-10096-0] [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] [Accepted: 05/23/2023] [Indexed: 06/10/2023]
Abstract
Cholangiocarcinoma (CCA), a bile duct cancer with a high mortality rate, has a poor prognosis due to its highly invasive and drug-resistant phenotypes. More effective and selective therapies are urgently needed. Bacteriocins are broad-spectrum antimicrobial peptides/proteins produced by bacterial strains to compete with other bacteria. Recent studies have reported that bacteriocins exhibit anticancer properties against various cancer cell lines with minimal toxicity toward normal cells. In this study, two types of recombinant bacteriocins, rhamnosin from probiotic Lacticaseibacillus rhamnosus and lysostaphin from Staphylococcus simulans, were highly produced in Escherichia coli and subsequently purified via immobilized-Ni2+ affinity chromatography. When their anticancer activity was investigated against CCA cell lines, both rhamnosin and lysostaphin were found capable of inhibiting the growth of CCA cell lines in a dose-dependent fashion but were less toxic toward a normal cholangiocyte cell line. Rhamnosin and lysostaphin as single treatments could suppress the growth of gemcitabine-resistant cell lines to the same extent as or more than they suppressed the parental counterparts. A combination of both bacteriocins more strongly inhibited growth and enhanced cell apoptosis in both parental and gemcitabine-resistant cells partly through the increased expression of the proapoptotic genes BAX, and caspase-3, -8, and -9. In conclusion, this is the first report to demonstrate an anticancer property of rhamnosin and lysostaphin. Using these bacteriocins as single agents or in combination would be effective against drug-resistant CCA.
Collapse
Affiliation(s)
- Kankamol Kerdkumthong
- Division of Health and Applied Sciences, Biochemistry Graduate Program, Faculty of Science, Prince of Songkla University, Hatyai, 90110, Songkhla, Thailand
| | - Wannarat Chanket
- Division of Health and Applied Sciences, Biochemistry Graduate Program, Faculty of Science, Prince of Songkla University, Hatyai, 90110, Songkhla, Thailand
- Graduate Program in Molecular Medicine, Faculty of Science, Mahidol University, Bangkok, 10400, Thailand
| | - Phanthipha Runsaeng
- Division of Health and Applied Sciences, Biochemistry Graduate Program, Faculty of Science, Prince of Songkla University, Hatyai, 90110, Songkhla, Thailand
| | - Sutthipong Nanarong
- Division of Health and Applied Sciences, Biochemistry Graduate Program, Faculty of Science, Prince of Songkla University, Hatyai, 90110, Songkhla, Thailand
| | - Kawinnath Songsurin
- Division of Health and Applied Sciences, Biochemistry Graduate Program, Faculty of Science, Prince of Songkla University, Hatyai, 90110, Songkhla, Thailand
| | - Phonprapavee Tantimetta
- Division of Health and Applied Sciences, Biochemistry Graduate Program, Faculty of Science, Prince of Songkla University, Hatyai, 90110, Songkhla, Thailand
| | - Chanan Angsuthanasombat
- Bacterial Toxin Research Innovation Laboratory, Institute of Molecular Biosciences, Mahidol University, Salaya Campus, Nakornpathom, 73170, Thailand
- Department of Biochemistry, School of Medicine, Tzu Chi University, Hualien, 97004, Taiwan
- Graduate Program in Immunology, Department of Immunology, Faculty of Medicine Siriraj Hospital, Mahidol University, Bangkok, 10700, Thailand
| | - Aratee Aroonkesorn
- Division of Health and Applied Sciences, Biochemistry Graduate Program, Faculty of Science, Prince of Songkla University, Hatyai, 90110, Songkhla, Thailand
| | - Sumalee Obchoei
- Division of Health and Applied Sciences, Biochemistry Graduate Program, Faculty of Science, Prince of Songkla University, Hatyai, 90110, Songkhla, Thailand.
| |
Collapse
|
12
|
Yarahmadi A, Zare M, Aghayari M, Afkhami H, Jafari GA. Therapeutic bacteria and viruses to combat cancer: double-edged sword in cancer therapy: new insights for future. Cell Commun Signal 2024; 22:239. [PMID: 38654309 PMCID: PMC11040964 DOI: 10.1186/s12964-024-01622-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2023] [Accepted: 04/17/2024] [Indexed: 04/25/2024] Open
Abstract
Cancer, ranked as the second leading cause of mortality worldwide, leads to the death of approximately seven million people annually, establishing itself as one of the most significant health challenges globally. The discovery and identification of new anti-cancer drugs that kill or inactivate cancer cells without harming normal and healthy cells and reduce adverse effects on the immune system is a potential challenge in medicine and a fundamental goal in Many studies. Therapeutic bacteria and viruses have become a dual-faceted instrument in cancer therapy. They provide a promising avenue for cancer treatment, but at the same time, they also create significant obstacles and complications that contribute to cancer growth and development. This review article explores the role of bacteria and viruses in cancer treatment, examining their potential benefits and drawbacks. By amalgamating established knowledge and perspectives, this review offers an in-depth examination of the present research landscape within this domain and identifies avenues for future investigation.
Collapse
Affiliation(s)
- Aref Yarahmadi
- Department of Biology, Khorramabad Branch, Islamic Azad University, Khorramabad, Iran
| | - Mitra Zare
- Department of Microbiology, Faculty of Sciences, Kerman Branch, Islamic Azad University, Kerman, Iran
| | - Masoomeh Aghayari
- Department of Microbiology, Faculty of Sciences, Urmia Branch, Islamic Azad University, Urmia, Iran
| | - Hamed Afkhami
- Nervous System Stem Cells Research Center, Semnan University of Medical Sciences, Semnan, Iran.
- Cellular and Molecular Research Center, Qom University of Medical Sciences, Qom, Iran.
- Department of Medical Microbiology, Faculty of Medicine, Shahed University, Tehran, Iran.
| | - Gholam Ali Jafari
- Cellular and Molecular Research Center, Qom University of Medical Sciences, Qom, Iran.
| |
Collapse
|
13
|
Wen-Tao C, Zhang YY, Qiang Q, Zou P, Xu Y, Sun C, Badar IH. Characterizations and molecular docking mechanism of the interactions between peptide FDGDF (Phe-Asp-Gly-Asp-Phe) and SOD enzyme. Heliyon 2024; 10:e24515. [PMID: 38293362 PMCID: PMC10826827 DOI: 10.1016/j.heliyon.2024.e24515] [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: 12/30/2022] [Revised: 01/05/2024] [Accepted: 01/10/2024] [Indexed: 02/01/2024] Open
Abstract
In this study, we investigated the antioxidant properties of dry-cured beef crude peptide (BPH) at different storage periods. The combination characteristics of different concentrations of Phe-Asp-Gly-Asp-Phe (FDGDF) and superoxide dismutase (SOD) at different temperatures were analyzed by ultraviolet-visible spectroscopy, fluorescence spectroscopy, and FT-IR spectroscopy, combined with the detection of a SOD activity detection box. It was found that FDGDF could improve the activity of SOD by changing its secondary structure. Bonds were formed at O32/O40/O52 using quantum chemical simulation calculations, and the Fukui index was higher than that of most atoms, indicating that these atoms were more likely to participate in the reaction. SPR biological force analysis showed that FDGDF and SOD were in a fast binding and dissociation mode. This study revealed the theoretical basis for studying the antioxidant mechanism of dry-cured beef and provided ideas for developing new dry-cured beef products.
Collapse
Affiliation(s)
- C.H.E.N. Wen-Tao
- School of Biological and Food Engineering, Changzhou University, Changzhou, Jiangsu, 213164, China
| | - Ying-Yang Zhang
- School of Biological and Food Engineering, Changzhou University, Changzhou, Jiangsu, 213164, China
| | - Qiang Qiang
- Changzhou Wujin No. 3 People's Hospital Changzhou, Jiangsu,150030, China
| | - Ping Zou
- School of Biological and Food Engineering, Changzhou University, Changzhou, Jiangsu, 213164, China
| | - Ying Xu
- School of Biological and Food Engineering, Changzhou University, Changzhou, Jiangsu, 213164, China
| | - Chengjun Sun
- School of Biological and Food Engineering, Changzhou University, Changzhou, Jiangsu, 213164, China
| | - Iftikhar Hussain Badar
- Department of Meat Science and Technology, University of Veterinary and Animal Sciences, Lahore 54000, Pakistan
- College of Food Science, Northeast Agricultural University, Harbin, Heilongjiang 150030, China
| |
Collapse
|
14
|
Gu Q, Yan J, Lou Y, Zhang Z, Li Y, Zhu Z, Liu M, Wu D, Liang Y, Pu J, Zhao X, Xiao H, Li P. Bacteriocins: Curial guardians of gastrointestinal tract. Compr Rev Food Sci Food Saf 2024; 23:e13292. [PMID: 38284593 DOI: 10.1111/1541-4337.13292] [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: 08/30/2023] [Revised: 12/05/2023] [Accepted: 12/14/2023] [Indexed: 01/30/2024]
Abstract
The human gastrointestinal (GI) tract microbiome secretes various metabolites that play pivotal roles in maintaining host physiological balance and influencing disease progression. Among these metabolites, bacteriocins-small, heat-stable peptides synthesized by ribosomes-are notably prevalent in the GI region. Their multifaceted benefits have garnered significant interest in the scientific community. This review comprehensively explores the methods for mining bacteriocins (traditional separation and purification, bioinformatics, and artificial intelligence), their effects on the stomach and intestines, and their complex bioactive mechanisms. These mechanisms include flora regulation, biological barrier restoration, and intervention in epithelial cell pathways. By detailing each well-documented bacteriocin, we reveal the diverse ways in which bacteriocins interact with the GI environment. Moreover, the future research direction is prospected. By further studying the function and interaction of intestinal bacteriocins, we can discover new pharmacological targets and develop drugs targeting intestinal bacteriocins to regulate and improve human health. It provides innovative ideas and infinite possibilities for further exploration, development, and utilization of bacteriocins. The inevitable fact is that the continuously exploration of bacteriocins is sure to bring the promising future for demic GI health understanding and interference strategy.
Collapse
Affiliation(s)
- Qing Gu
- Key Laboratory for Food Microbial Technology of Zhejiang Province, College of Food Science and Biotechnology, Zhejiang Gongshang University, Hangzhou, Zhejiang, People's Republic of China
| | - Jiaqian Yan
- Key Laboratory for Food Microbial Technology of Zhejiang Province, College of Food Science and Biotechnology, Zhejiang Gongshang University, Hangzhou, Zhejiang, People's Republic of China
| | - Yeqing Lou
- Key Laboratory for Food Microbial Technology of Zhejiang Province, College of Food Science and Biotechnology, Zhejiang Gongshang University, Hangzhou, Zhejiang, People's Republic of China
| | - Zihao Zhang
- Key Laboratory for Food Microbial Technology of Zhejiang Province, College of Food Science and Biotechnology, Zhejiang Gongshang University, Hangzhou, Zhejiang, People's Republic of China
| | - Yonglu Li
- Key Laboratory for Food Microbial Technology of Zhejiang Province, College of Food Science and Biotechnology, Zhejiang Gongshang University, Hangzhou, Zhejiang, People's Republic of China
| | - Zichun Zhu
- Key Laboratory for Food Microbial Technology of Zhejiang Province, College of Food Science and Biotechnology, Zhejiang Gongshang University, Hangzhou, Zhejiang, People's Republic of China
| | - Manman Liu
- Key Laboratory for Food Microbial Technology of Zhejiang Province, College of Food Science and Biotechnology, Zhejiang Gongshang University, Hangzhou, Zhejiang, People's Republic of China
| | - Danli Wu
- Key Laboratory for Food Microbial Technology of Zhejiang Province, College of Food Science and Biotechnology, Zhejiang Gongshang University, Hangzhou, Zhejiang, People's Republic of China
| | - Ying Liang
- Key Laboratory for Food Microbial Technology of Zhejiang Province, College of Food Science and Biotechnology, Zhejiang Gongshang University, Hangzhou, Zhejiang, People's Republic of China
| | - Jiaqian Pu
- Key Laboratory for Food Microbial Technology of Zhejiang Province, College of Food Science and Biotechnology, Zhejiang Gongshang University, Hangzhou, Zhejiang, People's Republic of China
| | - Xiaodan Zhao
- Key Laboratory for Food Microbial Technology of Zhejiang Province, College of Food Science and Biotechnology, Zhejiang Gongshang University, Hangzhou, Zhejiang, People's Republic of China
| | - Hang Xiao
- Department of Food Science, University of Massachusetts Amherst, Amherst, Massachusetts, USA
| | - Ping Li
- Key Laboratory for Food Microbial Technology of Zhejiang Province, College of Food Science and Biotechnology, Zhejiang Gongshang University, Hangzhou, Zhejiang, People's Republic of China
| |
Collapse
|
15
|
Kaźmierczak-Siedlecka K, Bulman N, Ulasiński P, Sobocki BK, Połom K, Marano L, Kalinowski L, Skonieczna-Żydecka K. Pharmacomicrobiomics of cell-cycle specific anti-cancer drugs - is it a new perspective for personalized treatment of cancer patients? Gut Microbes 2023; 15:2281017. [PMID: 37985748 PMCID: PMC10730203 DOI: 10.1080/19490976.2023.2281017] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/03/2023] [Accepted: 11/05/2023] [Indexed: 11/22/2023] Open
Abstract
Intestinal bacteria are equipped with an enzyme apparatus that is involved in the active biotransformation of xenobiotics, including drugs. Pharmacomicrobiomics, a new area of pharmacology, analyses interactions between bacteria and xenobiotics. However, there is another side to the coin. Pharmacotherapeutic agents can significantly modify the microbiota, which consequently affects their efficacy. In this review, we comprehensively gathered scientific evidence on the interplay between anticancer therapies and gut microbes. We also underlined how such interactions might impact the host response to a given therapy. We discuss the possibility of modulating the gut microbiota to increase the effectiveness/decrease the incidence of adverse events during tumor therapy. The anticipation of the future brings new evidence that gut microbiota is a target of interest to increase the efficacy of therapy.
Collapse
Affiliation(s)
- Karolina Kaźmierczak-Siedlecka
- Department of Medical Laboratory Diagnostics - Fahrenheit Biobank BBMRI.pl, Medical University of Gdansk, Gdańsk, Poland
| | - Nikola Bulman
- Department of Medical Laboratory Diagnostics - Fahrenheit Biobank BBMRI.pl, Medical University of Gdansk, Gdańsk, Poland
| | - Paweł Ulasiński
- Unit of Surgery with Unit of Oncological Surgery in Koscierzyna, Kościerzyna, Poland
| | - Bartosz Kamil Sobocki
- Department of Oncology and Radiotherapy, Medical University of Gdansk, Gdańsk, Poland
| | - Karol Połom
- Academy of Medical and Social Applied Sciences, Elbląg, Poland
| | - Luigi Marano
- Academy of Medical and Social Applied Sciences, Elbląg, Poland
| | - Leszek Kalinowski
- Department of Medical Laboratory Diagnostics - Fahrenheit Biobank BBMRI.pl, Medical University of Gdansk, Gdańsk, Poland
- BioTechMed Centre/Department of Mechanics of Materials and Structures, Gdansk University of Technology, Gdansk, Poland
| | | |
Collapse
|
16
|
Brango-Vanegas J, Leite ML, de Oliveira KBS, da Cunha NB, Franco OL. From exploring cancer and virus targets to discovering active peptides through mRNA display. Pharmacol Ther 2023; 252:108559. [PMID: 37952905 DOI: 10.1016/j.pharmthera.2023.108559] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2023] [Revised: 10/26/2023] [Accepted: 11/06/2023] [Indexed: 11/14/2023]
Abstract
During carcinogenesis, neoplastic cells accumulate mutations in genes important for cellular homeostasis, producing defective proteins. Viral infections occur when viral capsid proteins bind to the host cell receptor, allowing the virus to enter the cells. In both cases, proteins play important roles in cancer development and viral infection, so these targets can be exploited to develop alternative treatments. mRNA display technology is a very powerful tool for the development of peptides capable of acting on specific targets in neoplastic cells or on viral capsid proteins. mRNA display technology allows the selection and evolution of peptides with desired functional properties from libraries of many nucleic acid variants. Among other advantages of this technology, the use of flexizymes allows the production of peptides with unnatural amino acid residues, which can enhance the activity of these molecules. From target immobilization, peptides with greater specificity for the targets of interest are generated during the selection rounds. Herein, we will explore the use of mRNA display technology for the development of active peptides after successive rounds of selection, using proteins present in neoplastic cells and viral particles as targets.
Collapse
Affiliation(s)
- José Brango-Vanegas
- Centro de Análises Proteômicas e Bioquímicas, Programa de Pós-Graduação em Ciências Genômicas e Biotecnologia, Universidade Católica de Brasília, Brasília, DF, Brazil; S-inova Biotech, Programa de Pós-Graduação em Biotecnologia, Universidade Católica Dom Bosco, Campo Grande, MS, Brazil
| | - Michel Lopes Leite
- Departamento de Biologia Molecular, Instituto de Ciências Biológicas, Universidade de Brasília, Campus Darcy Ribeiro, Brasília, DF, Brazil
| | - Kamila Botelho Sampaio de Oliveira
- Centro de Análises Proteômicas e Bioquímicas, Programa de Pós-Graduação em Ciências Genômicas e Biotecnologia, Universidade Católica de Brasília, Brasília, DF, Brazil; S-inova Biotech, Programa de Pós-Graduação em Biotecnologia, Universidade Católica Dom Bosco, Campo Grande, MS, Brazil
| | - Nicolau Brito da Cunha
- Universidade de Brasília, Faculdade de Agronomia e Medicina Veterinária, Campus Darcy Ribeiro, Brasília, DF, Brazil
| | - Octávio Luiz Franco
- Centro de Análises Proteômicas e Bioquímicas, Programa de Pós-Graduação em Ciências Genômicas e Biotecnologia, Universidade Católica de Brasília, Brasília, DF, Brazil; S-inova Biotech, Programa de Pós-Graduação em Biotecnologia, Universidade Católica Dom Bosco, Campo Grande, MS, Brazil.
| |
Collapse
|
17
|
Din SRU, Saeed S, Khan SU, Arbi FM, Xuefang G, Zhong M. Bacteria-driven cancer therapy: Exploring advancements and challenges. Crit Rev Oncol Hematol 2023; 191:104141. [PMID: 37742883 DOI: 10.1016/j.critrevonc.2023.104141] [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: 08/08/2023] [Revised: 09/15/2023] [Accepted: 09/18/2023] [Indexed: 09/26/2023] Open
Abstract
Cancer, a serious fatal disease caused by the uncontrolled growth of cells, is the biggest challenge flagging around medicine and health fields. Conventionally, various treatments-based strategies such as radiotherapy, chemotherapy, and alternative cancer therapies possess drugs that cannot reach the cancerous tissues and make them toxic to noncancerous cells. Cancer immunotherapy has made outstanding achievements in reducing the chances of cancer. Our considerable attention towards cancer-directed immune responses and the mechanisms behind which immune cells kill cancer cells have progressively been helpful in the advancement of new therapies. Among them, bacteria-based cancer immunotherapy has achieved much more attention due to smart and robust mechanisms in activating the host anti-tumor response. Moreover, bacterial-based therapy can be utilized as a single monotherapy or in combination with multiple anticancer immunotherapies to accelerate productive clinical results. Herein, we comprehensively reviewed recent advancements, challenges, and future perspectives in developing bacterial-based cancer immunotherapies.
Collapse
Affiliation(s)
- Syed Riaz Ud Din
- Department of Microbiology, College of Basic Medical Sciences, Dalian Medical University, Dalian 116044, China
| | - Sumbul Saeed
- School of Environment and Science, Griffith University, Nathan, QLD 4111, Australia
| | - Shahid Ullah Khan
- Integrative Science Center of Germplasm Creation in Western China (CHONGQING) Science City and Southwest University, College of Agronomy and Biotechnology, Southwest University, Beibei, Chongqing 400715, China; Engineering Research Center of South Upland Agriculture, Ministry of Education, Chongqing 400715, China; Women Medical and Dental College, Khyber Medical University, Peshawar, KPK 22020, Pakistan
| | - Fawad Mueen Arbi
- Quaid-e-Azam Medical College, Bahawalpur, Punjab 63100, Pakistan
| | - Guo Xuefang
- Department of Medical Microbiology, Dalian Medical University, Dalian 116044, China
| | - Mintao Zhong
- Department of Microbiology, College of Basic Medical Sciences, Dalian Medical University, Dalian 116044, China.
| |
Collapse
|
18
|
Chukwudulue UM, Barger N, Dubovis M, Luzzatto Knaan T. Natural Products and Pharmacological Properties of Symbiotic Bacillota (Firmicutes) of Marine Macroalgae. Mar Drugs 2023; 21:569. [PMID: 37999393 PMCID: PMC10672036 DOI: 10.3390/md21110569] [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: 10/06/2023] [Revised: 10/24/2023] [Accepted: 10/25/2023] [Indexed: 11/25/2023] Open
Abstract
The shift from the terrestrial to the marine environment to discover natural products has given rise to novel bioactive compounds, some of which have been approved for human medicine. However, the ocean, which makes up nearly three-quarters of the Earth's surface, contains macro- and microorganisms whose natural products are yet to be explored. Among these underexplored marine organisms are macroalgae and their symbiotic microbes, such as Bacillota, a phylum of mostly Gram-positive bacteria previously known as Firmicutes. Macroalgae-associated Bacillota often produce chemical compounds that protect them and their hosts from competitive and harmful rivals. Here, we summarised the natural products made by macroalgae-associated Bacillota and their pharmacological properties. We discovered that these Bacillota are efficient producers of novel biologically active molecules. However, only a few macroalgae had been investigated for chemical constituents of their Bacillota: nine brown, five red and one green algae. Thus, Bacillota, especially from the marine habitat, should be investigated for potential pharmaceutical leads. Moreover, additional diverse biological assays for the isolated molecules of macroalgae Bacillota should be implemented to expand their bioactivity profiles, as only antibacterial properties were tested for most compounds.
Collapse
Affiliation(s)
| | | | | | - Tal Luzzatto Knaan
- Department of Marine Biology, The Charney School of Marine Sciences, University of Haifa, Mount Carmel, Haifa 103301, Israel; (U.M.C.); (N.B.); (M.D.)
| |
Collapse
|
19
|
Haghighatafshar H, Golestani Eimani B, Moazamian E, Amani J. Cytotoxic and apoptotic effects of chemically synthesized silver nanoparticles loaded with recombinant Staphylococcus LukS-PV toxin. J Biotechnol 2023; 373:42-48. [PMID: 37421980 DOI: 10.1016/j.jbiotec.2023.07.001] [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: 09/06/2022] [Revised: 06/13/2023] [Accepted: 07/02/2023] [Indexed: 07/10/2023]
Abstract
Chronic myeloid leukemia (CML) accounts for approximately 15% of leukemias. LukS-PV, a Panton-Valentine leucocidin (PVL) component, is secreted by Staphylococcus aureus. Silver nanoparticles have increasingly been used for different purposes, most notably for drug delivery and anticancer agents. In this work, the cytotoxicity effect of recombinant LukS-PV protein, chemically synthesized AgNPs, and recombinant LukS-PV protein-loaded silver nanoparticles was investigated on human Chronic myeloid leukemia K562 cells and human normal embryonic kidney HEK293 cells. Cell apoptosis was investigated by staining with Annexin V/propidium iodide. The recombinant LukS-PV protein-loaded silver nanoparticles exhibited dose-dependent cytotoxicity and induced apoptosis in the K562 cells but had little effect on normal HEK293 cells. After 24 h of exposure to recombinant LukS-PV protein-loaded silver nanoparticles (IC50 concentration), flow cytometry showed that 31.17% of K562 cells were apoptotic. These results indicate that recombinant LukS-PV protein-loaded silver nanoparticles maybe are a potential chemotherapeutic agent candidate against K562 cells. Hence, silver nanoparticles could be used as drug carriers for toxin release to cancer cells.
Collapse
Affiliation(s)
- Hafizeh Haghighatafshar
- Department of Microbiology, Faculty of Sciences, Agriculture and Modern Technology, Shiraz Branch, Islamic Azad University, Shiraz, Iran
| | | | - Elham Moazamian
- Department of Microbiology, Faculty of Sciences, Agriculture and Modern Technology, Shiraz Branch, Islamic Azad University, Shiraz, Iran
| | - Jafar Amani
- Applied Microbiology Research Center, Systems Biology and Poisonings Institute, Baqiyatallah University of Medical Sciences, Tehran, Iran
| |
Collapse
|
20
|
Gostaviceanu A, Gavrilaş S, Copolovici L, Copolovici DM. Membrane-Active Peptides and Their Potential Biomedical Application. Pharmaceutics 2023; 15:2091. [PMID: 37631305 PMCID: PMC10459175 DOI: 10.3390/pharmaceutics15082091] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2023] [Revised: 07/24/2023] [Accepted: 08/03/2023] [Indexed: 08/27/2023] Open
Abstract
Membrane-active peptides (MAPs) possess unique properties that make them valuable tools for studying membrane structure and function and promising candidates for therapeutic applications. This review paper provides an overview of the fundamental aspects of MAPs, focusing on their membrane interaction mechanisms and potential applications. MAPs exhibit various structural features, including amphipathic structures and specific amino acid residues, enabling selective interaction with multiple membranes. Their mechanisms of action involve disrupting lipid bilayers through different pathways, depending on peptide properties and membrane composition. The therapeutic potential of MAPs is significant. They have demonstrated antimicrobial activity against bacteria and fungi, making them promising alternatives to conventional antibiotics. MAPs can selectively target cancer cells and induce apoptosis, opening new avenues in cancer therapeutics. Additionally, MAPs serve as drug delivery vectors, facilitating the transport of therapeutic cargoes across cell membranes. They represent a fascinating class of biomolecules with significant potential in basic research and clinical applications. Understanding their mechanisms of action and designing peptides with enhanced selectivity and efficacy will further expand their utility in diverse fields. Exploring MAPs holds promise for developing novel therapeutic strategies against infections, cancer, and drug delivery challenges.
Collapse
Affiliation(s)
- Andreea Gostaviceanu
- Faculty of Food Engineering, Tourism and Environmental Protection, and Institute for Research, Development and Innovation in Technical and Natural Sciences, Aurel Vlaicu University, Elena Drăgoi St., No. 2, 310330 Arad, Romania; (A.G.); (S.G.); (L.C.)
- Biomedical Sciences Doctoral School, University of Oradea, University St., No. 1, 410087 Oradea, Romania
| | - Simona Gavrilaş
- Faculty of Food Engineering, Tourism and Environmental Protection, and Institute for Research, Development and Innovation in Technical and Natural Sciences, Aurel Vlaicu University, Elena Drăgoi St., No. 2, 310330 Arad, Romania; (A.G.); (S.G.); (L.C.)
| | - Lucian Copolovici
- Faculty of Food Engineering, Tourism and Environmental Protection, and Institute for Research, Development and Innovation in Technical and Natural Sciences, Aurel Vlaicu University, Elena Drăgoi St., No. 2, 310330 Arad, Romania; (A.G.); (S.G.); (L.C.)
| | - Dana Maria Copolovici
- Faculty of Food Engineering, Tourism and Environmental Protection, and Institute for Research, Development and Innovation in Technical and Natural Sciences, Aurel Vlaicu University, Elena Drăgoi St., No. 2, 310330 Arad, Romania; (A.G.); (S.G.); (L.C.)
| |
Collapse
|
21
|
Thoda C, Touraki M. Probiotic-Derived Bioactive Compounds in Colorectal Cancer Treatment. Microorganisms 2023; 11:1898. [PMID: 37630458 PMCID: PMC10456921 DOI: 10.3390/microorganisms11081898] [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: 06/30/2023] [Revised: 07/14/2023] [Accepted: 07/24/2023] [Indexed: 08/27/2023] Open
Abstract
Colorectal cancer (CRC) is a multifactorial disease with increased morbidity and mortality rates globally. Despite advanced chemotherapeutic approaches for the treatment of CRC, low survival rates due to the regular occurrence of drug resistance and deleterious side effects render the need for alternative anticancer agents imperative. Accumulating evidence supports that gut microbiota imbalance precedes the establishment of carcinogenesis, subsequently contributing to cancer progression and response to anticancer therapy. Manipulation of the gut microbiota composition via the administration of probiotic-derived bioactive compounds has gradually attained the interest of scientific communities as a novel therapeutic strategy for CRC. These compounds encompass miscellaneous metabolic secreted products of probiotics, including bacteriocins, short-chain fatty acids (SCFAs), lactate, exopolysaccharides (EPSs), biosurfactants, and bacterial peptides, with profound anti-inflammatory and antiproliferative properties. This review provides a classification of postbiotic types and a comprehensive summary of the current state of research on their biological role against CRC. It also describes how their intricate interaction with the gut microbiota regulates the proper function of the intestinal barrier, thus eliminating gut dysbiosis and CRC development. Finally, it discusses the future perspectives in precision-medicine approaches as well as the challenges of their synthesis and optimization of administration in clinical studies.
Collapse
Affiliation(s)
| | - Maria Touraki
- Laboratory of General Biology, Department of Genetics, Development and Molecular Biology, School of Biology, Faculty of Sciences, Aristotle University of Thessaloniki, 54 124 Thessaloniki, Greece;
| |
Collapse
|
22
|
Nagpal S, Mande SS. Environmental insults and compensative responses: when microbiome meets cancer. Discov Oncol 2023; 14:130. [PMID: 37453005 DOI: 10.1007/s12672-023-00745-9] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/12/2023] [Accepted: 07/04/2023] [Indexed: 07/18/2023] Open
Abstract
Tumor microenvironment has recently been ascribed a new hallmark-the polymorphic microbiome. Accumulating evidence regarding the tissue specific territories of tumor-microbiome have opened new and interesting avenues. A pertinent question is regarding the functional consequence of the interface between host-microbiome and cancer. Given microbial communities have predominantly been explored through an ecological perspective, it is important that the foundational aspects of ecological stress and the fight to 'survive and thrive' are accounted for tumor-micro(b)environment as well. Building on existing evidence and classical microbial ecology, here we attempt to characterize the ecological stresses and the compensative responses of the microorganisms inside the tumor microenvironment. What insults would microbes experience inside the cancer jungle? How would they respond to these insults? How the interplay of stress and microbial quest for survival would influence the fate of tumor? This work asks these questions and tries to describe this underdiscussed ecological interface of the tumor and its microbiota. It is hoped that a larger scientific thought on the importance of microbial competition sensing vis-à-vis tumor-microenvironment would be stimulated.
Collapse
Affiliation(s)
- Sunil Nagpal
- TCS Research, Tata Consultancy Services Ltd, Pune, 411013, India.
- CSIR-Institute of Genomics and Integrative Biology (CSIR-IGIB), New Delhi, 110025, India.
- Academy of Scientific and Innovative Research (AcSIR), Ghaziabad, 201002, India.
| | - Sharmila S Mande
- TCS Research, Tata Consultancy Services Ltd, Pune, 411013, India.
| |
Collapse
|
23
|
Ojha P, Kar NP, Behera HT, Parija M, Nayak S, Singh S, Patra AK, Sahoo KK. Independent antioxidant and anticancer properties of a novel thermostable lysozyme isolated from Bacillus paralicheniformis: in silico and in vitro studies. 3 Biotech 2023; 13:240. [PMID: 37337524 PMCID: PMC10276796 DOI: 10.1007/s13205-023-03653-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2022] [Accepted: 05/28/2023] [Indexed: 06/21/2023] Open
Abstract
In this study, we evaluated the independent anticancer properties of a novel heat-stable lysozyme derived from the thermophilic bacterium Bacillus paralicheniformis (BplzC) to identify potential alternative therapies to address the suboptimal outcomes of current cancer treatments. Using the String 10.5 database, an in-silico protein-protein interaction study predicted that BplzC was a strong functional partner of cytochrome c, indicating a potential role in cancer cell apoptosis. Further, the HDOCK server predicted that BplzC strongly bound to cell death receptors, such as cytokines FAS receptor, leading to activation of cytochrome c and subsequent apoptosis in the cancer cell line. In vitro assays demonstrated uniform apoptotic activity of BplzC against various cancer cell lines, while showing no apoptotic activity against normal non-cancer cell lines. And showing no apoptotic activity against normal non-cancer cell lines suggested a very specific mode of action and without any adverse side effects. Additionally, BplzC exhibited ROS scavenging activity and reducing ability comparable to ascorbic acid, and significantly accelerated HEK293 cell migration. Our findings suggest that BplzC has specific cytotoxic effects on cancer cells and may be a valuable natural source of antioxidants for future use in the nutritional and pharmaceutical sectors.
Collapse
Affiliation(s)
- Purusottam Ojha
- Department of Botany, Ravenshaw University, Cuttack, Odisha 753003 India
- Imgenex India Pvt. Ltd., E5 Infocity, Chandka Industrial Estate, KIIT Post Office, Bhubaneswar, Odisha 751024 India
| | - Narayani Prasad Kar
- Department of Biological Sciences, North Carolina State University, Raleigh, NC USA
| | - Himadri Tanaya Behera
- Department of Biotechnology, KIIT Deemed to be University, Bhubaneswar, Odisha India
| | - Manaswini Parija
- Department of Botany, Ravenshaw University, Cuttack, Odisha 753003 India
| | - Shreenath Nayak
- Imgenex India Pvt. Ltd., E5 Infocity, Chandka Industrial Estate, KIIT Post Office, Bhubaneswar, Odisha 751024 India
| | - Sujay Singh
- Imgenex India Pvt. Ltd., E5 Infocity, Chandka Industrial Estate, KIIT Post Office, Bhubaneswar, Odisha 751024 India
| | - Ashok Kumar Patra
- Imgenex India Pvt. Ltd., E5 Infocity, Chandka Industrial Estate, KIIT Post Office, Bhubaneswar, Odisha 751024 India
| | - Khirod Kumar Sahoo
- Department of Botany, Ravenshaw University, Cuttack, Odisha 753003 India
| |
Collapse
|
24
|
Ikryannikova LN, Gorokhovets NV, Belykh DA, Kurbatov LK, Zamyatnin AA. Bacterial Therapy of Cancer: A Way to the Dustbin of History or to the Medicine of the Future? Int J Mol Sci 2023; 24:ijms24119726. [PMID: 37298677 DOI: 10.3390/ijms24119726] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2023] [Revised: 05/23/2023] [Accepted: 05/30/2023] [Indexed: 06/12/2023] Open
Abstract
Bacteria are the constant companions of the human body throughout its life and even after its death. The history of a human disease such as cancer and the history of microorganisms, particularly bacteria, are believed to closely intertwined. This review was conceived to highlight the attempts of scientists from ancient times to the present day to discover the relationship between bacteria and the emergence or development of tumors in the human body. Challenges and achievements of 21st century science in forcing bacteria to serve for cancer treatment are considered. The future possibilities of bacterial cancer therapy, including the creation of bacterial microrobots, or "bacteriobots", are also discussed.
Collapse
Affiliation(s)
- Larisa N Ikryannikova
- Institute of Molecular Medicine, Sechenov First Moscow State Medical University, Trubetskaya 8/2, 119991 Moscow, Russia
| | - Neonila V Gorokhovets
- Institute of Molecular Medicine, Sechenov First Moscow State Medical University, Trubetskaya 8/2, 119991 Moscow, Russia
| | - Darya A Belykh
- Institute of Molecular Medicine, Sechenov First Moscow State Medical University, Trubetskaya 8/2, 119991 Moscow, Russia
| | - Leonid K Kurbatov
- Orekhovich Research Institute of Biomedical Chemistry, Pogodinskaya 10/8, 119991 Moscow, Russia
| | - Andrey A Zamyatnin
- Institute of Molecular Medicine, Sechenov First Moscow State Medical University, Trubetskaya 8/2, 119991 Moscow, Russia
- Faculty of Bioengineering and Bioinformatics, Lomonosov Moscow State University, Leninskie Gory 1/73, 119234 Moscow, Russia
- Belozersky Research Institute of Physico-Chemical Biology, Lomonosov Moscow State University, Leninskie Gory 1/40, 119992 Moscow, Russia
- Scientific Center for Translation Medicine, Sirius University of Science and Technology, 1 Olympic Ave, 354340 Sochi, Russia
| |
Collapse
|
25
|
Soundararajan S, Selvakumar J, Maria Joseph ZM, Gopinath Y, Saravanan V, Santhanam R. Investigating the modulatory effects of Moringa oleifera on the gut microbiota of chicken model through metagenomic approach. Front Vet Sci 2023; 10:1153769. [PMID: 37323848 PMCID: PMC10267347 DOI: 10.3389/fvets.2023.1153769] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2023] [Accepted: 04/17/2023] [Indexed: 06/17/2023] Open
Abstract
Introduction This study aimed to assess the effects of supplementing chicken feed with Moringa oleifera leaf powder, a phytobiotic, on the gastrointestinal microbiota. The objective was to examine the microbial changes induced by the supplementation. Methods A total of 40, one-day-old chickens were fed their basal diet for 42 days and then divided into two groups: SG1 (basal diet) and SG2 (basal diet + 10 g/kg Moringa oleifera leaf powder). Metagenomics analysis was conducted to analyze operational taxonomic units (OTUs), species annotation, and biodiversity. Additionally, 16S rRNA sequencing was performed for molecular characterization of isolated gut bacteria, identified as Enterococcus faecium. The isolated bacteria were tested for essential metabolites, demonstrating antibacterial, antioxidant, and anticancer activities. Results and discussion The analysis revealed variations in the microbial composition between the control group (SG1) and the M. oleifera-treated group (SG2). SG2 showed a 47% increase in Bacteroides and a 30% decrease in Firmicutes, Proteobacteria, Actinobacteria, and Tenericutes compared to SG1. TM7 bacteria were observed exclusively in the M. oleifera-treated group. These findings suggest that Moringa oleifera leaf powder acts as a modulator that enhances chicken gut microbiota, promoting the colonization of beneficial bacteria. PICRUSt analysis supported these findings, showing increased carbohydrate and lipid metabolism in the M.oleifera-treated gut microbiota. Conclusion This study indicates that supplementing chicken feed with Moringa oleifera leaf powder as a phytobiotic enhances the gut microbiota in chicken models, potentially improving overall health. The observed changes in bacterial composition, increased presence of Bacteroides, and exclusive presence of TM7 bacteria suggest a positive modulation of microbial balance. The essential metabolites from isolated Enterococcus faecium bacteria further support the potential benefits of Moringa oleifera supplementation.
Collapse
Affiliation(s)
- Sowmiya Soundararajan
- Department of Biotechnology and Bioinformatics, Bishop Heber College (Autonomous), Affiliated With Bharathidasan University, Tiruchirappalli, Tamil Nadu, India
| | - Jasmine Selvakumar
- Department of Biotechnology and Bioinformatics, Bishop Heber College (Autonomous), Affiliated With Bharathidasan University, Tiruchirappalli, Tamil Nadu, India
| | - Zion Mercy Maria Joseph
- Department of Biotechnology and Bioinformatics, Bishop Heber College (Autonomous), Affiliated With Bharathidasan University, Tiruchirappalli, Tamil Nadu, India
| | - Yuvapriya Gopinath
- Department of Biotechnology and Bioinformatics, Bishop Heber College (Autonomous), Affiliated With Bharathidasan University, Tiruchirappalli, Tamil Nadu, India
| | - Vaishali Saravanan
- Department of Biotechnology and Bioinformatics, Bishop Heber College (Autonomous), Affiliated With Bharathidasan University, Tiruchirappalli, Tamil Nadu, India
| | - Rameshkumar Santhanam
- Faculty of Science and Marine Environment, Universiti Malaysia Terengganu, Kuala Nerus, Terengganu, Malaysia
| |
Collapse
|
26
|
Thakker DP, Narayanan R. Arginine deiminase produced by lactic acid bacteria as a potent anti-cancer drug. Med Oncol 2023; 40:175. [PMID: 37171497 DOI: 10.1007/s12032-023-02043-4] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2023] [Accepted: 04/30/2023] [Indexed: 05/13/2023]
Abstract
Bacterial-based cancer immunotherapy has recently gained widespread attention due to its exceptional mechanism of rich pathogen-associated molecular patterns in anti-cancer immune responses. Contrary to conventional cancer therapies such as surgery, chemotherapy, radiation and phototherapy, bacteria-based cancer immunotherapy has the unique ability to suppress cancer by selectively accumulating and growing in tumours. In the view of this, several bacterial strains are being used for the treatment of cancer. Of which, lactic acid bacteria are a powerful, albeit still inadequately understood bacteria that possess a wide source of bioactive chemicals. Lactic acid bacteria metabolites, such as bacteriocins, short-chain fatty acids, exopolysaccharides show antitumour property. Amino acid pathways, which have lately been focussed as a new strategy to cancer therapy, are key element of the adaptability and dysregulation of metabolic pathways identified in proliferation of tumour cells. Arginine metabolism, in particular, has been shown to be critical for cancer therapy. As a result, better understanding of arginine metabolism in LAB and cancer cells could lead to new cancer therapeutic targets. This review will outline current advances in the interaction of arginine metabolism with cancer therapy and propose an arginine deiminase expression system to combat cancer more effectively.
Collapse
Affiliation(s)
- Darshali P Thakker
- Department of Genetic Engineering, College of Engineering & Technology, SRM Institute of Science and Technology, SRM Nagar, Kattankulathur, 603203, Tamil Nadu, India
| | - Rajnish Narayanan
- Department of Genetic Engineering, College of Engineering & Technology, SRM Institute of Science and Technology, SRM Nagar, Kattankulathur, 603203, Tamil Nadu, India.
| |
Collapse
|
27
|
Proteins and their functionalization for finding therapeutic avenues in cancer: Current status and future prospective. Biochim Biophys Acta Rev Cancer 2023; 1878:188862. [PMID: 36791920 DOI: 10.1016/j.bbcan.2023.188862] [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: 06/24/2022] [Revised: 01/13/2023] [Accepted: 01/13/2023] [Indexed: 02/15/2023]
Abstract
Despite the remarkable advancement in the health care sector, cancer remains the second most fatal disease globally. The existing conventional cancer treatments primarily include chemotherapy, which has been associated with little to severe side effects, and radiotherapy, which is usually expensive. To overcome these problems, target-specific nanocarriers have been explored for delivering chemo drugs. However, recent reports on using a few proteins having anticancer activity and further use of them as drug carriers have generated tremendous attention for furthering the research towards cancer therapy. Biomolecules, especially proteins, have emerged as suitable alternatives in cancer treatment due to multiple favourable properties including biocompatibility, biodegradability, and structural flexibility for easy surface functionalization. Several in vitro and in vivo studies have reported that various proteins derived from animal, plant, and bacterial species, demonstrated strong cytotoxic and antiproliferative properties against malignant cells in native and their different structural conformations. Moreover, surface tunable properties of these proteins help to bind a range of anticancer drugs and target ligands, thus making them efficient delivery agents in cancer therapy. Here, we discuss various proteins obtained from common exogenous sources and how they transform into effective anticancer agents. We also comprehensively discuss the tumor-killing mechanisms of different dietary proteins such as bovine α-lactalbumin, hen egg-white lysozyme, and their conjugates. We also articulate how protein nanostructures can be used as carriers for delivering cancer drugs and theranostics, and strategies to be adopted for improving their in vivo delivery and targeting. We further discuss the FDA-approved protein-based anticancer formulations along with those in different phases of clinical trials.
Collapse
|
28
|
Metagenomics Analysis of Breast Microbiome Highlights the Abundance of Rothia Genus in Tumor Tissues. J Pers Med 2023; 13:jpm13030450. [PMID: 36983633 PMCID: PMC10053322 DOI: 10.3390/jpm13030450] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2023] [Revised: 02/20/2023] [Accepted: 02/21/2023] [Indexed: 03/05/2023] Open
Abstract
Breast cancer is one of the main global priorities in terms of public health. It remains the most frequent cancer in women and is the leading cause of their death. The human microbiome plays various roles in maintaining health by ensuring a dynamic balance with the host or in the appearance of various pathologies including breast cancer. In this study, we performed an analysis of bacterial signature differences between tumor and adjacent tissues of breast cancer patients in Morocco. Using 16S rRNA gene sequencing, we observed that adjacent tissue contained a much higher percentage of the Gammaproteobacteria class (35.7%) while tumor tissue was characterized by a higher percentage of Bacilli and Actinobacteria classes, with about 18.8% and 17.2% average abundance, respectively. Analysis of tumor subtype revealed enrichment of genus Sphingomonodas in TNBC while Sphingomonodas was predominant in HER2. The LEfSe and the genus level heatmap analysis revealed a higher abundance of the Rothia genus in tumor tissues. The identified microbial communities can therefore serve as potential biomarkers for prognosis and diagnosis, while also helping to develop new strategies for the treatment of breast cancer patients.
Collapse
|
29
|
Gándara Z, Rubio N, Castillo RR. Delivery of Therapeutic Biopolymers Employing Silica-Based Nanosystems. Pharmaceutics 2023; 15:pharmaceutics15020351. [PMID: 36839672 PMCID: PMC9963032 DOI: 10.3390/pharmaceutics15020351] [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: 12/01/2022] [Revised: 01/15/2023] [Accepted: 01/17/2023] [Indexed: 01/22/2023] Open
Abstract
The use of nanoparticles is crucial for the development of a new generation of nanodevices for clinical applications. Silica-based nanoparticles can be tailored with a wide range of functional biopolymers with unique physicochemical properties thus providing several advantages: (1) limitation of interparticle interaction, (2) preservation of cargo and particle integrity, (3) reduction of immune response, (4) additional therapeutic effects and (5) cell targeting. Therefore, the engineering of advanced functional coatings is of utmost importance to enhance the biocompatibility of existing biomaterials. Herein we will focus on the most recent advances reported on the delivery and therapeutic use of silica-based nanoparticles containing biopolymers (proteins, nucleotides, and polysaccharides) with proven biological effects.
Collapse
Affiliation(s)
- Zoila Gándara
- Departamento de Química Orgánica y Química Inorgánica, Universidad de Alcalá, 28805 Alcalá de Henares, Spain
- Instituto de Investigación Química “Andrés M. del Río” (IQAR), Universidad de Alcalá, 28805 Alcalá de Henares, Spain
- Correspondence: (Z.G.); (N.R.); (R.R.C.)
| | - Noelia Rubio
- Departamento de Química Orgánica y Química Inorgánica, Universidad de Alcalá, 28805 Alcalá de Henares, Spain
- Instituto de Investigación Química “Andrés M. del Río” (IQAR), Universidad de Alcalá, 28805 Alcalá de Henares, Spain
- Correspondence: (Z.G.); (N.R.); (R.R.C.)
| | - Rafael R. Castillo
- Departamento de Química Orgánica y Química Inorgánica, Universidad de Alcalá, 28805 Alcalá de Henares, Spain
- Instituto de Investigación Química “Andrés M. del Río” (IQAR), Universidad de Alcalá, 28805 Alcalá de Henares, Spain
- Correspondence: (Z.G.); (N.R.); (R.R.C.)
| |
Collapse
|
30
|
Hu J, Jiang W, Zuo J, Shi D, Chen X, Yang X, Zhang W, Ma L, Liu Z, Xing Q. Structural basis of bacterial effector protein azurin targeting tumor suppressor p53 and inhibiting its ubiquitination. Commun Biol 2023; 6:59. [PMID: 36650277 PMCID: PMC9845241 DOI: 10.1038/s42003-023-04458-1] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2022] [Accepted: 01/10/2023] [Indexed: 01/19/2023] Open
Abstract
Tumor suppressor p53 prevents tumorigenesis by promoting cell cycle arrest and apoptosis through transcriptional regulation. Dysfunction of p53 occurs frequently in human cancers. Thus, p53 becomes one of the most promising targets for anticancer treatment. A bacterial effector protein azurin triggers tumor suppression by stabilizing p53 and elevating its basal level. However, the structural and mechanistic basis of azurin-mediated tumor suppression remains elusive. Here we report the atomic details of azurin-mediated p53 stabilization by combining X-ray crystallography with nuclear magnetic resonance. Structural and mutagenic analysis reveals that the p28 region of azurin, which corresponds to a therapeutic peptide, significantly contributes to p53 binding. This binding stabilizes p53 by disrupting COP1-mediated p53 ubiquitination and degradation. Using the structure-based design, we obtain several affinity-enhancing mutants that enable amplifying the effect of azurin-induced apoptosis. Our findings highlight how the structure of the azurin-p53 complex can be leveraged to design azurin derivatives for cancer therapy.
Collapse
Affiliation(s)
- Jianjian Hu
- grid.35155.370000 0004 1790 4137National Key Laboratory of Crop Genetic Improvement, Hubei Hongshan Laboratory, Huazhong Agricultural University, Wuhan, 430070 China
| | - Wenxue Jiang
- grid.34418.3a0000 0001 0727 9022State Key Laboratory of Biocatalysis and Enzyme Engineering, College of Life Sciences, Hubei University, Wuhan, 430074 China
| | - Jiaqi Zuo
- grid.35155.370000 0004 1790 4137National Key Laboratory of Crop Genetic Improvement, Hubei Hongshan Laboratory, Huazhong Agricultural University, Wuhan, 430070 China
| | - Dujuan Shi
- grid.34418.3a0000 0001 0727 9022State Key Laboratory of Biocatalysis and Enzyme Engineering, College of Life Sciences, Hubei University, Wuhan, 430074 China
| | - Xiaoqi Chen
- grid.34418.3a0000 0001 0727 9022State Key Laboratory of Biocatalysis and Enzyme Engineering, College of Life Sciences, Hubei University, Wuhan, 430074 China
| | - Xiao Yang
- grid.34418.3a0000 0001 0727 9022State Key Laboratory of Biocatalysis and Enzyme Engineering, College of Life Sciences, Hubei University, Wuhan, 430074 China
| | - Wenhui Zhang
- grid.35155.370000 0004 1790 4137National Key Laboratory of Crop Genetic Improvement, Hubei Hongshan Laboratory, Huazhong Agricultural University, Wuhan, 430070 China
| | - Lixin Ma
- grid.34418.3a0000 0001 0727 9022State Key Laboratory of Biocatalysis and Enzyme Engineering, College of Life Sciences, Hubei University, Wuhan, 430074 China
| | - Zhu Liu
- grid.34418.3a0000 0001 0727 9022State Key Laboratory of Biocatalysis and Enzyme Engineering, College of Life Sciences, Hubei University, Wuhan, 430074 China
| | - Qiong Xing
- grid.34418.3a0000 0001 0727 9022State Key Laboratory of Biocatalysis and Enzyme Engineering, College of Life Sciences, Hubei University, Wuhan, 430074 China
| |
Collapse
|
31
|
Yaghoubi A, Movaqar A, Asgharzadeh F, Derakhshan M, Ghazvini K, Hasanian SM, Avan A, Mostafapour A, Khazaei M, Soleimanpour S. Anticancer activity of Pseudomonas aeruginosa derived peptide with iRGD in colon cancer therapy. IRANIAN JOURNAL OF BASIC MEDICAL SCIENCES 2023; 26:768-776. [PMID: 37396945 PMCID: PMC10311979 DOI: 10.22038/ijbms.2023.68331.14913] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Figures] [Subscribe] [Scholar Register] [Received: 10/09/2022] [Accepted: 03/01/2023] [Indexed: 07/04/2023]
Abstract
Objectives Colon cancer is well-known as a life-threatening disease. Since the current treatment modalities for this type of cancer are powerful yet face some limitations, finding novel treatments is required to achieve better outcomes with fewer side effects. Here we investigated the therapeutic potential of Azurin-p28 alone or along with iRGD (Ac-CRGDKGPDC-amide) as a tumor-penetrating peptide and 5-fluorouracil (5-FU) for colon cancer. Materials and Methods Inhibitory effect of p28 with or without iRGD/5-FU was studied in CT26 and HT29, as well as the xenograft animal model of cancer. The effect of p28 alone or along with iRGD/5-FU on cell migration, apoptotic activity, and cell cycle of the cell lines was assessed. Level of the BAX and BCL2 genes, tumor suppressor genes [(p53 and collagen type-Iα1 (COL1A1), collagen type-Iα2 (COL1A2)] were assessed by quantitative RT-PCR. Results These findings show that using p28 with or without iRGD and 5-FU raised the level of p53 and BAX but decreased BCL2, compared with control and 5-FU groups in tissues of the tumor, which result in raising the apoptosis. Conclusion It seems that p28 may be used as a new therapeutic approach in colon cancer therapy that can enhance the anti-tumor effect of 5-FU.
Collapse
Affiliation(s)
- Atieh Yaghoubi
- Department of Microbiology and Virology, Faculty of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
- Student Research Committee, Faculty of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Aref Movaqar
- Department of Microbiology and Virology, Faculty of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Fereshteh Asgharzadeh
- Student Research Committee, Faculty of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
- Department of Physiology, Faculty of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Mohammad Derakhshan
- Department of Microbiology and Virology, Faculty of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Kiarash Ghazvini
- Department of Microbiology and Virology, Faculty of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Seyed Mahdi Hasanian
- Department of Medical Biochemistry, Faculty of Medicine, Mashhad University of Medical, Sciences, Mashhad, Iran
| | - Amir Avan
- Department of Medical Genetics and Molecular Medicine, Faculty of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Asma Mostafapour
- Department of Biology, Mashhad Branch, Islamic Azad University, Mashhad, Iran
| | - Majid Khazaei
- Department of Physiology, Faculty of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Saman Soleimanpour
- Department of Microbiology and Virology, Faculty of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
| |
Collapse
|
32
|
Yadav M, Eswari JS. Opportunistic Challenges of Computer-aided Drug Discovery of Lipopeptides: New Insights for Large Molecule Therapeutics. Avicenna J Med Biotechnol 2023; 15:3-13. [PMID: 36789119 PMCID: PMC9895984 DOI: 10.18502/ajmb.v15i1.11419] [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: 04/04/2022] [Accepted: 08/27/2022] [Indexed: 12/27/2022] Open
Abstract
Computer-aided drug designing is a promising approach to defeating the dry pipeline of drug discovery. It aims at reduced experimental efforts with cost-effectiveness. Naturally occurring large molecules with molecular weight higher than 500 Dalton such as cationic peptides, cyclic peptides, glycopeptides and lipopeptides are a few examples of large molecules which have successful applications as the broad spectrum antibacterial, anticancer, antiviral, antifungal and antithrombotic drugs. Utilization of microbial metabolites as potential drug candidates incur cost effectiveness through large scale production of such molecules rather than a synthetic approach. Computational studies on such compounds generate tremendous possibilities to develop novel leads with challenges to handle these complex molecules with available computational tools. The opportunities begin with the desired structural modifications in the parent drug molecule. Virtual modifications followed by molecular interaction studies at the target site through molecular modeling simulations and identification of structure-activity relationship models to develop more prominent and potential drug molecules. Lead optimization studies to develop novel compounds with increased specificity and reduced off targeting is a big challenge computationally for large molecules. Prediction of optimized pharmacokinetic properties facilitates development of a compound with lower toxicity as compared to the natural compounds. Generating the library of compounds and studies for target specificity and ADMET (Absorption, Distribution, Metabolism, Excretion and Toxicity) for large molecules are laborious and incur huge cost and chemical wastage through in-vitro methods. Hence, computational methods need to be explored to develop novel compounds from natural large molecules with higher specificity. This review article is focusing on possible challenges and opportunities in the pathway of computer-aided drug discovery of large molecule therapeutics.
Collapse
Affiliation(s)
- Manisha Yadav
- Department of Biotechnology, National Institute of Technology Raipur, C.G., India
| | - J. Satya Eswari
- Department of Biotechnology, National Institute of Technology Raipur, C.G., India
| |
Collapse
|
33
|
Rawangkan A, Wongsirisin P, Pook-In G, Siriphap A, Yosboonruang A, Kiddee A, Chuerduangphui J, Reukngam N, Duangjai A, Saokaew S, Praphasawat R. Dinactin: A New Antitumor Antibiotic with Cell Cycle Progression and Cancer Stemness Inhibiting Activities in Lung Cancer. Antibiotics (Basel) 2022; 11:antibiotics11121845. [PMID: 36551502 PMCID: PMC9774622 DOI: 10.3390/antibiotics11121845] [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: 11/30/2022] [Revised: 12/13/2022] [Accepted: 12/16/2022] [Indexed: 12/23/2022] Open
Abstract
Lung cancer, especially non-small cell lung cancer (NSCLC), is one of the most complex diseases, despite the existence of effective treatments such as chemotherapy and immunotherapy. Since cancer stem cells (CSCs) are responsible for chemo- and radio-resistance, metastasis, and cancer recurrence, finding new therapeutic targets for CSCs is critical. Dinactin is a natural secondary metabolite produced by microorganisms. Recently, dinactin has been revealed as a promising antitumor antibiotic via various mechanisms. However, the evidence relating to cell cycle progression regulation is constrained, and effects on cancer stemness have not been elucidated. Therefore, the aim of this study is to evaluate the new function of dinactin in anti-NSCLC proliferation, focusing on cell cycle progression and cancer stemness properties in Lu99 and A549 cells. Flow cytometry and immunoblotting analyses revealed that 0.1-1 µM of dinactin suppresses cell growth through induction of the G0/G1 phase associated with down-regulation of cyclins A, B, and D3, and cdk2 protein expression. The tumor-sphere forming capacity was used to assess the effect of dinactin on the cancer stemness potential in NSCLC cells. At a concentration of 1 nM, dinactin reduced both the number and size of the tumor-spheres. The quantitative RT-PCR analyses indicated that dinactin suppressed sphere formation by significantly reducing expression of CSC markers (i.e., ALDH1A1, Nanog, Oct4, and Sox2) in Lu99 cells. Consequently, dinactin could be a promising strategy for NSCLC therapy targeting CSCs.
Collapse
Affiliation(s)
- Anchalee Rawangkan
- Division of Microbiology and Parasitology, School of Medical Sciences, University of Phayao, Phayao 56000, Thailand
- UNIt of Excellence on Clinical Outcomes Research and IntegratioN (UNICORN), School of Pharmaceutical Sciences, University of Phayao, Phayao 56000, Thailand
| | - Pattama Wongsirisin
- Department of Medical Services, National Cancer Institute, Bangkok 10400, Thailand
| | - Grissana Pook-In
- Division of Microbiology and Parasitology, School of Medical Sciences, University of Phayao, Phayao 56000, Thailand
| | - Achiraya Siriphap
- Division of Microbiology and Parasitology, School of Medical Sciences, University of Phayao, Phayao 56000, Thailand
| | - Atchariya Yosboonruang
- Division of Microbiology and Parasitology, School of Medical Sciences, University of Phayao, Phayao 56000, Thailand
| | - Anong Kiddee
- Division of Microbiology and Parasitology, School of Medical Sciences, University of Phayao, Phayao 56000, Thailand
| | | | - Nanthawan Reukngam
- Laboratory of Organic Synthesis, Chulabhorn Research Institute, Bangkok 10210, Thailand
| | - Acharaporn Duangjai
- Division of Physiology, School of Medical Sciences, University of Phayao, Phayao 56000, Thailand
| | - Surasak Saokaew
- UNIt of Excellence on Clinical Outcomes Research and IntegratioN (UNICORN), School of Pharmaceutical Sciences, University of Phayao, Phayao 56000, Thailand
- Division of Social and Administrative Pharmacy, Department of Pharmaceutical Care, School of Pharmaceutical Sciences, University of Phayao, Phayao 56000, Thailand
| | - Ratsada Praphasawat
- Department of Pathology, School of Medicine, University of Phayao, Phayao 56000, Thailand
- Correspondence: ; Tel.: +66-54466666 (ext. 3824) or +66-86-926-2448
| |
Collapse
|
34
|
Lee JH, Lee SB, Kim H, Shin JM, Yoon M, An HS, Han JW. Anticancer Activity of Mannose-Specific Lectin, BPL2, from Marine Green Alga Bryopsis plumosa. Mar Drugs 2022; 20:md20120776. [PMID: 36547923 PMCID: PMC9788543 DOI: 10.3390/md20120776] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2022] [Revised: 12/05/2022] [Accepted: 12/07/2022] [Indexed: 12/15/2022] Open
Abstract
Lectin is a carbohydrate-binding protein that recognizes specific cells by binding to cell-surface polysaccharides. Tumor cells generally show various glycosylation patterns, making them distinguishable from non-cancerous cells. Consequently, lectin has been suggested as a good anticancer agent. Herein, the anticancer activity of Bryopsis plumosa lectins (BPL1, BPL2, and BPL3) was screened and tested against lung cancer cell lines (A549, H460, and H1299). BPL2 showed high anticancer activity compared to BPL1 and BPL3. Cell viability was dependent on BPL2 concentration and incubation time. The IC50 value for lung cancer cells was 50 μg/mL after 24 h of incubation in BPL2 containing medium; however, BPL2 (50 μg/mL) showed weak toxicity in non-cancerous cells (MRC5). BPL2 affected cancer cell growth while non-cancerous cells were less affected. Further, BPL2 (20 μg/mL) inhibited cancer cell invasion and migration (rates were ˂20%). BPL2 induced the downregulation of epithelial-to-mesenchymal transition-related genes (Zeb1, vimentin, and Twist). Co-treatment with BPL2 and gefitinib (10 μg/mL and 10 μM, respectively) showed a synergistic effect compared with monotherapy. BPL2 or gefitinib monotherapy resulted in approximately 90% and 70% cell viability, respectively, with concomitant treatment showing 40% cell viability. Overall, BPL2 can be considered a good candidate for development into an anticancer agent.
Collapse
|
35
|
Zhou C, Peng D, Liao B, Jia R, Wu F. ACP_MS: prediction of anticancer peptides based on feature extraction. Brief Bioinform 2022; 23:6793775. [PMID: 36326080 DOI: 10.1093/bib/bbac462] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2022] [Revised: 09/10/2022] [Accepted: 09/27/2022] [Indexed: 11/06/2022] Open
Abstract
Anticancer peptides (ACPs) are bioactive peptides with antitumor activity and have become the most promising drugs in the treatment of cancer. Therefore, the accurate prediction of ACPs is of great significance to the research of cancer diseases. In the paper, we developed a more efficient prediction model called ACP_MS. Firstly, the monoMonoKGap method is used to extract the characteristic of anticancer peptide sequences and form the digital features. Then, the AdaBoost model is used to select the most discriminating features from the digital features. Finally, a stochastic gradient descent algorithm is introduced to identify anticancer peptide sequences. We adopt 7-fold cross-validation and independent test set validation, and the final accuracy of the main dataset reached 92.653% and 91.597%, respectively. The accuracy of the alternate dataset reached 98.678% and 98.317%, respectively. Compared with other advanced prediction models, the ACP_MS model improves the identification ability of anticancer peptide sequences. The data of this model can be downloaded from the public website for free https://github.com/Zhoucaimao1998/Zc.
Collapse
Affiliation(s)
- Caimao Zhou
- Key Laboratory of Computational Science and Application of Hainan Province, Haikou, China.,Key Laboratory of Data Science and Intelligence Education, Hainan Normal University, Ministry of Education, Haikou, China.,School of Mathematics and Statistics, Hainan Normal University, Haikou, China
| | - Dejun Peng
- Key Laboratory of Computational Science and Application of Hainan Province, Haikou, China.,Key Laboratory of Data Science and Intelligence Education, Hainan Normal University, Ministry of Education, Haikou, China.,School of Mathematics and Statistics, Hainan Normal University, Haikou, China
| | - Bo Liao
- Key Laboratory of Computational Science and Application of Hainan Province, Haikou, China.,Key Laboratory of Data Science and Intelligence Education, Hainan Normal University, Ministry of Education, Haikou, China.,School of Mathematics and Statistics, Hainan Normal University, Haikou, China
| | - Ranran Jia
- Key Laboratory of Computational Science and Application of Hainan Province, Haikou, China.,Key Laboratory of Data Science and Intelligence Education, Hainan Normal University, Ministry of Education, Haikou, China.,School of Mathematics and Statistics, Hainan Normal University, Haikou, China
| | - Fangxiang Wu
- Key Laboratory of Computational Science and Application of Hainan Province, Haikou, China.,Key Laboratory of Data Science and Intelligence Education, Hainan Normal University, Ministry of Education, Haikou, China.,School of Mathematics and Statistics, Hainan Normal University, Haikou, China
| |
Collapse
|
36
|
Garbacz K. Anticancer activity of lactic acid bacteria. Semin Cancer Biol 2022; 86:356-366. [PMID: 34995799 DOI: 10.1016/j.semcancer.2021.12.013] [Citation(s) in RCA: 49] [Impact Index Per Article: 24.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2021] [Revised: 12/29/2021] [Accepted: 12/30/2021] [Indexed: 01/27/2023]
Abstract
Lactic acid bacteria (LAB), a group of Gram-positive microorganisms naturally occurring in fermented food products and used as probiotics, have been gaining the interest of researchers for years. LAB are potent, albeit still not wholly understood, source of bioactive compounds with various functions and activity. Metabolites of LAB, among others, short-chain fatty acids, exopolysaccharides and bacteriocins have promising anticancer potential. Research on the interactions between the bioactive metabolites of LAB and immune mechanisms demonstrated that these substances could exert a strong immunomodulatory effect, which would explain their vast therapeutic potential. The anticancer activity of LAB was confirmed both in vitro and in animal models against cancer cells from various malignancies. LAB inhibit tumor growth through various mechanisms, including antiproliferative activity, induction of apoptosis, cell cycle arrest, as well as through antimutagenic, antiangiogenic and anti-inflammatory effects. The aim of this review was to summarize the most recent data about the anticancer activity of LAB, with particular emphasis on the most promising bioactive compounds with potential clinical application.
Collapse
Affiliation(s)
- Katarzyna Garbacz
- Department of Oral Microbiology, Medical Faculty, Medical University of Gdansk, 25 Dębowa Str., 80-204, Gdansk, Poland.
| |
Collapse
|
37
|
Recent advances in microbial toxin-related strategies to combat cancer. Semin Cancer Biol 2022; 86:753-768. [PMID: 34271147 DOI: 10.1016/j.semcancer.2021.07.007] [Citation(s) in RCA: 26] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2021] [Revised: 07/01/2021] [Accepted: 07/09/2021] [Indexed: 02/08/2023]
Abstract
It is a major concern to treat cancer successfully, due to the distinctive pathophysiology of cancer cells and the gradual manifestation of resistance. Specific action, adverse effects and development of resistance has prompted the urgent requirement of exploring alternative anti-tumour treatment therapies. The naturally derived microbial toxins as a therapy against cancer cells are a promisingly new dimension. Various important microbial toxins such as Diphtheria toxin, Vibrio cholera toxin, Aflatoxin, Patulin, Cryptophycin-55, Chlorella are derived from several bacterial, fungal and algal species. These agents act on different biotargets such as inhibition of protein synthesis, reduction in cell growth, regulation of cell cycle and many cellular processes. Bacterial toxins produce actions primarily by targeting protein moieties and some immunomodulation and few acts through DNA. Fungal toxins appear to have more DNA damaging activity and affect the cell cycle. Algal toxins produce alteration in mitochondrial phosphorylation. In conclusion, microbial toxins and their metabolites appear to have a great potential to provide a promising option for the treatment and management to combat cancer.
Collapse
|
38
|
Diwan D, Cheng L, Usmani Z, Sharma M, Holden N, Willoughby N, Sangwan N, Baadhe RR, Liu C, Gupta VK. Microbial cancer therapeutics: A promising approach. Semin Cancer Biol 2022; 86:931-950. [PMID: 33979677 DOI: 10.1016/j.semcancer.2021.05.003] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2021] [Revised: 04/24/2021] [Accepted: 05/04/2021] [Indexed: 01/27/2023]
Abstract
The success of conventional cancer therapeutics is hindered by associated dreadful side-effects of antibiotic resistance and the dearth of antitumor drugs' selectivity and specificity. Hence, the conceptual evolution of anti-cancerous therapeutic agents that selectively target cancer cells without impacting the healthy cells or tissues, has led to a new wave of scientific interest in microbial-derived bioactive molecules. Such strategic solutions may pave the way to surmount the shortcomings of conventional therapies and raise the potential and hope for the cure of wide range of cancer in a selective manner. This review aims to provide a comprehensive summary of anti-carcinogenic properties and underlying mechanisms of bioactive molecules of microbial origin, and discuss the current challenges and effective therapeutic application of combinatorial strategies to attain minimal systemic side-effects.
Collapse
Affiliation(s)
- Deepti Diwan
- Washington University, School of Medicine, Saint Louis, MO, USA
| | - Lei Cheng
- Department of Pulmonary, Shanghai Chest Hospital, Shanghai Jiao Tong University, Shanghai, 230032, China
| | - Zeba Usmani
- Department of Chemistry and Biotechnology, Tallinn University of Technology, 12618, Tallinn, Estonia
| | - Minaxi Sharma
- Department of Food Technology, Akal College of Agriculture, Eternal University, Baru Sahib, Himachal Pradesh, 173101, India
| | - Nicola Holden
- Centre for Safe and Improved Food, Scotland's Rural College (SRUC), Kings Buildings, West Mains Road, Edinburgh, EH9 3JG, UK
| | - Nicholas Willoughby
- Institute of Biological Chemistry, Biophysics and Bioengineering, School of Engineering and Physical Sciences, Heriot-Watt University, Edinburgh, EH14 4AS, UK
| | - Neelam Sangwan
- Department of Biochemistry, Central University of Haryana, Mahendergarh, Haryana, 123031, India
| | - Rama Raju Baadhe
- Department of Biotechnology, National Institute of Technology, Warangal, Telangana, 506004, India
| | - Chenchen Liu
- Department of Gastric Surgery, Fudan University Shanghai Cancer Center, Shanghai, 200032, China; Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, 200032, China.
| | - Vijai Kumar Gupta
- Centre for Safe and Improved Food, Scotland's Rural College (SRUC), Kings Buildings, West Mains Road, Edinburgh, EH9 3JG, UK; Biorefining and Advanced Materials Research Center, Scotland's Rural College (SRUC), Kings Buildings, West Mains Road, Edinburgh, EH9 3JG, UK.
| |
Collapse
|
39
|
The Chemotherapeutic Potentials of Compounds Isolated from the Plant, Marine, Fungus, and Microorganism: Their Mechanism of Action and Prospects. J Trop Med 2022; 2022:5919453. [PMID: 36263439 PMCID: PMC9576449 DOI: 10.1155/2022/5919453] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2022] [Accepted: 09/10/2022] [Indexed: 12/02/2022] Open
Abstract
Research on natural products mainly focuses on developing a suitable drug to treat human disease. There has been a sharp increase in the development of drugs from natural products. Most of the drugs that are available are from the terrestrial origin. Marine natural products are less explored. Oceans are considered as a vast ecosystem with a wide variety of living organisms and natural products that are unexplored. Large numbers of antitumor drugs are from natural sources such as plants, marine, and microorganisms. 80% new chemical entities that were launched over the past 60 decades were from a natural source. In this article, the anticancer potential from the natural source such as plants, fungi, microorganisms, marine, and endophytes has been reviewed. Emphasis is given on the compound from the marine, plant, and of bacterial origin. Finally, we consider the future and how we might achieve better sustainability to alleviate human cancer suffering while having fewer side effects, more efficacies, and causing less harm than the present treatments.
Collapse
|
40
|
Bacteriocins as Potential Therapeutic Approaches in the Treatment of Various Cancers: A Review of In Vitro Studies. Cancers (Basel) 2022; 14:cancers14194758. [PMID: 36230679 PMCID: PMC9563265 DOI: 10.3390/cancers14194758] [Citation(s) in RCA: 15] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2022] [Revised: 09/23/2022] [Accepted: 09/25/2022] [Indexed: 11/17/2022] Open
Abstract
Simple Summary Current cancer treatment strategies such as surgery, chemotherapy, and radiotherapy, have significant drawbacks. There is a need for a breakthrough approach to cancer treatment. Bacteriocin, an antimicrobial peptide, has shown several anticancer properties in vitro. Therefore, this article reviews the effect of bacteriocin on cancer cells and how bacteriocins affect cancer cells in vitro. This article aims to promote additional bacteriocin research, particularly in vivo studies, to fully understand the potential of bacteriocin as a cancer treatment agent. Abstract Cancer is regarded as one of the most common and leading causes of death. Despite the availability of conventional treatments against cancer cells, current treatments are not the optimal treatment for cancer as they possess the possibility of causing various unwanted side effects to the body. As a result, this prompts a search for an alternative treatment without exhibiting any additional side effects. One of the promising novel therapeutic candidates against cancer is an antimicrobial peptide produced by bacteria called bacteriocin. It is a non-toxic peptide that is reported to exhibit potency against cancer cell lines. Experimental studies have outlined the therapeutic potential of bacteriocin against various cancer cell lines. In this review article, the paper focuses on the various bacteriocins and their cytotoxic effects, mode of action and efficacies as therapeutic agents against various cancer cell lines.
Collapse
|
41
|
Marković KG, Grujović MŽ, Koraćević MG, Nikodijević DD, Milutinović MG, Semedo-Lemsaddek T, Djilas MD. Colicins and Microcins Produced by Enterobacteriaceae: Characterization, Mode of Action, and Putative Applications. INTERNATIONAL JOURNAL OF ENVIRONMENTAL RESEARCH AND PUBLIC HEALTH 2022; 19:11825. [PMID: 36142096 PMCID: PMC9517006 DOI: 10.3390/ijerph191811825] [Citation(s) in RCA: 17] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/26/2022] [Revised: 09/12/2022] [Accepted: 09/15/2022] [Indexed: 06/15/2023]
Abstract
Enterobacteriaceae are widely present in many environments related to humans, including the human body and the food that they consume, from both plant or animal origin. Hence, they are considered relevant members of the gastrointestinal tract microbiota. On the other hand, these bacteria are also recognized as putative pathogens, able to impair human health and, in food, they are considered indicators for the microbiological quality and hygiene status of a production process. Nevertheless, beneficial properties have also been associated with Enterobacteriaceae, such as the ability to synthesize peptides and proteins, which can have a role in the structure of microbial communities. Among these antimicrobial molecules, those with higher molecular mass are called colicins, while those with lower molecular mass are named microcins. In recent years, some studies show an emphasis on molecules that can help control the development of pathogens. However, not enough data are available on this subject, especially related to microcins. Hence, this review gathers and summarizes current knowledge on colicins and microcins, potential usage in the treatment of pathogen-associated diseases and cancer, as well as putative applications in food biotechnology.
Collapse
Affiliation(s)
- Katarina G. Marković
- Institute for Information Technologies, Department of Science, University of Kragujevac, Jovana Cvijića bb, 34000 Kragujevac, Serbia
| | - Mirjana Ž. Grujović
- Institute for Information Technologies, Department of Science, University of Kragujevac, Jovana Cvijića bb, 34000 Kragujevac, Serbia
| | - Maja G. Koraćević
- Innovation Center, University of Niš, 18000 Niš, Serbia
- Faculty of Medicine, Department of Pharmacy, University of Niš, 18000 Niš, Serbia
| | - Danijela D. Nikodijević
- Faculty of Science, Department of Biology and Ecology, University of Kragujevac, Radoja Domanovića 12, 34000 Kragujevac, Serbia
| | - Milena G. Milutinović
- Faculty of Science, Department of Biology and Ecology, University of Kragujevac, Radoja Domanovića 12, 34000 Kragujevac, Serbia
| | - Teresa Semedo-Lemsaddek
- CIISA—Centro de Investigação Interdisciplinar em Sanidade Animal, Faculdade de Medicina Veterinária, Universidade de Lisboa, Avenida da Universidade Técnica, 1300-477 Lisboa, Portugal
- Associate Laboratory for Animal and Veterinary Sciences (AL4AnimalS), 1300-477 Lisboa, Portugal
| | - Milan D. Djilas
- Institute for Public Health of Vojvodina, Futoška 121, 21000 Novi Sad, Serbia
| |
Collapse
|
42
|
Koo DJ, Sut TN, Tan SW, Yoon BK, Jackman JA. Biophysical Characterization of LTX-315 Anticancer Peptide Interactions with Model Membrane Platforms: Effect of Membrane Surface Charge. Int J Mol Sci 2022; 23:10558. [PMID: 36142470 PMCID: PMC9501188 DOI: 10.3390/ijms231810558] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2022] [Revised: 09/06/2022] [Accepted: 09/08/2022] [Indexed: 11/16/2022] Open
Abstract
LTX-315 is a clinical-stage, anticancer peptide therapeutic that disrupts cancer cell membranes. Existing mechanistic knowledge about LTX-315 has been obtained from cell-based biological assays, and there is an outstanding need to directly characterize the corresponding membrane-peptide interactions from a biophysical perspective. Herein, we investigated the membrane-disruptive properties of the LTX-315 peptide using three cell-membrane-mimicking membrane platforms on solid supports, namely the supported lipid bilayer, intact vesicle adlayer, and tethered lipid bilayer, in combination with quartz crystal microbalance-dissipation (QCM-D) and electrochemical impedance spectroscopy (EIS) measurements. The results showed that the cationic LTX-315 peptide selectively disrupted negatively charged phospholipid membranes to a greater extent than zwitterionic or positively charged phospholipid membranes, whereby electrostatic interactions were the main factor to influence peptide attachment and membrane curvature was a secondary factor. Of note, the EIS measurements showed that the LTX-315 peptide extensively and irreversibly permeabilized negatively charged, tethered lipid bilayers that contained high phosphatidylserine lipid levels representative of the outer leaflet of cancer cell membranes, while circular dichroism (CD) spectroscopy experiments indicated that the LTX-315 peptide was structureless and the corresponding membrane-disruptive interactions did not involve peptide conformational changes. Dynamic light scattering (DLS) measurements further verified that the LTX-315 peptide selectively caused irreversible disruption of negatively charged lipid vesicles. Together, our findings demonstrate that the LTX-315 peptide preferentially disrupts negatively charged phospholipid membranes in an irreversible manner, which reinforces its potential as an emerging cancer immunotherapy and offers a biophysical framework to guide future peptide engineering efforts.
Collapse
Affiliation(s)
- Dong Jun Koo
- School of Chemical Engineering and Translational Nanobioscience Research Center, Sungkyunkwan University, Suwon 16419, Korea
| | - Tun Naw Sut
- School of Chemical Engineering and Translational Nanobioscience Research Center, Sungkyunkwan University, Suwon 16419, Korea
| | - Sue Woon Tan
- School of Chemical Engineering and Translational Nanobioscience Research Center, Sungkyunkwan University, Suwon 16419, Korea
| | - Bo Kyeong Yoon
- School of Healthcare and Biomedical Engineering, Chonnam National University, Yeosu 59626, Korea
| | - Joshua A. Jackman
- School of Chemical Engineering and Translational Nanobioscience Research Center, Sungkyunkwan University, Suwon 16419, Korea
| |
Collapse
|
43
|
Babakanrad E, Mohammadian T, Esmaeili D, Behzadi P. Efficacy of the Apoptotic Activity of CpsA-CpsC-L-ACAN Fusion Peptide against HeLa Cell Line. MOLECULAR GENETICS, MICROBIOLOGY AND VIROLOGY 2022. [DOI: 10.3103/s089141682203003x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/26/2022]
|
44
|
Dicks LMT, Vermeulen W. Do Bacteria Provide an Alternative to Cancer Treatment and What Role Does Lactic Acid Bacteria Play? Microorganisms 2022; 10:microorganisms10091733. [PMID: 36144335 PMCID: PMC9501580 DOI: 10.3390/microorganisms10091733] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2022] [Revised: 08/17/2022] [Accepted: 08/26/2022] [Indexed: 11/16/2022] Open
Abstract
Cancer is one of the leading causes of mortality and morbidity worldwide. According to 2022 statistics from the World Health Organization (WHO), close to 10 million deaths have been reported in 2020 and it is estimated that the number of cancer cases world-wide could increase to 21.6 million by 2030. Breast, lung, thyroid, pancreatic, liver, prostate, bladder, kidney, pelvis, colon, and rectum cancers are the most prevalent. Each year, approximately 400,000 children develop cancer. Treatment between countries vary, but usually includes either surgery, radiotherapy, or chemotherapy. Modern treatments such as hormone-, immuno- and antibody-based therapies are becoming increasingly popular. Several recent reports have been published on toxins, antibiotics, bacteriocins, non-ribosomal peptides, polyketides, phenylpropanoids, phenylflavonoids, purine nucleosides, short chain fatty acids (SCFAs) and enzymes with anticancer properties. Most of these molecules target cancer cells in a selective manner, either directly or indirectly through specific pathways. This review discusses the role of bacteria, including lactic acid bacteria, and their metabolites in the treatment of cancer.
Collapse
|
45
|
Pattnaik S, Imchen M, Kumavath R, Prasad R, Busi S. Bioactive Microbial Metabolites in Cancer Therapeutics: Mining, Repurposing, and Their Molecular Targets. Curr Microbiol 2022; 79:300. [PMID: 36002695 DOI: 10.1007/s00284-022-02990-7] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2021] [Accepted: 08/01/2022] [Indexed: 11/30/2022]
Abstract
The persistence and resurgence of cancer, characterized by abnormal cell growth and differentiation, continues to be a serious public health concern critically affecting public health, social life, and the global economy. Hundreds of putative drug molecules of synthetic and natural origin were approved for anticancer therapy in the last few decades. Although conventional anticancer treatment strategies have promising aspects, several factors such as their limitations, drug resistance, and side effects associated with them demand more effort in repositioning or developing novel therapeutic regimens. The rich heritage of microbial bioactive components remains instrumental in providing novel avenues for cancer therapeutics. Actinobacteria, Firmicutes, and fungi have a plethora of bioactive compounds, which received attention for their efficacy in cancer treatment targeting different pathways responsible for abnormal cell growth and differentiation. Yet the full potential remains underexplored to date, and novel compounds from such microbes are reported regularly. In addition, the advent of computational tools has further augmented the mining of microbial secondary metabolites and identifying their molecular targets in cancer cells. Furthermore, the drug-repurposing strategy has facilitated the use of approved drugs of microbial origin in regulating cancer cell growth and progression. The wide diversity of microbial compounds, different mining approaches, and multiple modes of action warrant further investigations on the current status of microbial metabolites in cancer therapeutics. Hence, in this review, we have critically discussed the untapped potential of microbial products in mitigating cancer progression. The review also summarizes the impact of drug repurposing in cancer therapy and discusses the novel avenues for future therapeutic drug development against cancer.
Collapse
Affiliation(s)
- Subhaswaraj Pattnaik
- Department of Microbiology, School of Life Sciences, Pondicherry University, Puducherry, 605014, India.,Department of Biotechnology and Bioinformatics, Sambalpur University, Jyoti Vihar, Burla, Sambalpur, Odisha, 768019, India
| | - Madangchanok Imchen
- Department of Microbiology, School of Life Sciences, Pondicherry University, Puducherry, 605014, India.,Department of Genomic Science, School of Biological Sciences, Central University of Kerela, Kasaragod, Kerela, 671316, India
| | - Ranjith Kumavath
- Department of Genomic Science, School of Biological Sciences, Central University of Kerela, Kasaragod, Kerela, 671316, India
| | - Ram Prasad
- Department of Botany, School of Life Sciences, Mahatma Gandhi Central University, Motihari, Bihar, 845401, India.
| | - Siddhardha Busi
- Department of Microbiology, School of Life Sciences, Pondicherry University, Puducherry, 605014, India.
| |
Collapse
|
46
|
Singh N, Tapader R, Chatterjee S, Pal A, Pal A. Subtilisin from Bacillus amyloliquefaciens induces apoptosis in breast cancer cells through ubiquitin-proteasome-mediated tubulin degradation. Int J Biol Macromol 2022; 220:852-865. [PMID: 35985398 DOI: 10.1016/j.ijbiomac.2022.08.086] [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: 05/05/2022] [Revised: 07/20/2022] [Accepted: 08/11/2022] [Indexed: 11/05/2022]
Abstract
To search for novel proteases from environmental isolates which can induce apoptosis in cancer cells, we have purified subtilisin from Bacillus amyloliquefaciens and studied its anti-cancer properties. Subtilisin induced apoptosis in colon (HT29) and breast (MCF7) cancer cells but showed no effect on mouse peritoneal macrophages and normal breast cells (MCF10A). Western blot analysis showed that Bax, Bcl-2 level remained unchanged but tubulin level decreased significantly. Subtilisin does not induce the intrinsic pathway of apoptosis, rather it induced tubulin degradation in MCF-7 cells, whereas in normal cells (MCF-10A) tubulin degradation was not observed. Subtilisin activates ubiquitination and proteasomal-mediated tubulin degradation which was completely restored in presence of proteasome inhibitor MG-132. We further observed PARKIN, one of the known E3-ligase, is overexpressed and interacts with tubulin in subtilisin treated cells. Knockdown of PARKIN effectively downregulates ubiquitination and inhibits degradation of tubulin. PARKIN activation and tubulin degradation lead to ER-stress which in turn activates caspase-7 and PARP cleavage, thus guiding the subtilisin treated cells towards apoptosis. To our knowledge this is the first report of subtilisin induced apoptosis in cancer cells by proteasomal degradation of tubulin.
Collapse
Affiliation(s)
- Nanda Singh
- Division of Pathophysiology, ICMR-National Institute of Cholera and Enteric Diseases (NICED), Kolkata 700010, India
| | - Rima Tapader
- Division of Pathophysiology, ICMR-National Institute of Cholera and Enteric Diseases (NICED), Kolkata 700010, India
| | - Shruti Chatterjee
- Division of Biotechnology and Phycology, CSIR-Central Salt & Marine Chemical Research Institute, Bhavnagar 364002, India
| | - Ananda Pal
- Division of Clinical Medicine, ICMR-National Institute of Cholera and Enteric Diseases (NICED), Kolkata 700010, India
| | - Amit Pal
- Division of Pathophysiology, ICMR-National Institute of Cholera and Enteric Diseases (NICED), Kolkata 700010, India.
| |
Collapse
|
47
|
Haider T, Pandey V, Behera C, Kumar P, Gupta PN, Soni V. Nisin and nisin-loaded nanoparticles: a cytotoxicity investigation. Drug Dev Ind Pharm 2022; 48:310-321. [PMID: 35938875 DOI: 10.1080/03639045.2022.2111438] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/03/2022]
Abstract
OBJECTIVE Nisin is an antibacterial peptide with anticancer properties, but the main drawback is its rapid enzymatic degradation and limited permeation across the cell membrane. This research aims to to overcome these drawbacks by developing nisin-loaded nanoparticles with improved cytotoxic effects. SIGNIFICANCE PLGA nanoparticles are one of the most effective biodegradable and biocompatible drug delivery carriers. In the present study, nisin-loaded nanoparticles showed enhanced anticancer effects. METHODS NPN was prepared by a double emulsion solvent evaporation method and characterized for different parameters. The cytotoxic investigation of NPN was carried out on various cell lines, including A549, SW-620, HT-29, PC-3, MDA-MB-231, MCF-7, MiaPaca-2, and fR2 by sulforhodamine B (SRB) assay. Mechanistic investigation of cellular cytotoxicity was performed by using bright-field microscopy, DAPI staining, intracellular reactive oxygen species (ROS), changes in mitochondrial membrane potential (ΔΨm), and western blotting. A comparative cytotoxicity study of nisin and NPN was performed on normal breast epithelial cells (fR-2). RESULTS NPN showed spherical shape, 289.09 ± 3.63 nm particle size, and 63.37 ± 3.12% entrapment efficiency. NPN was more cytotoxic to the MDA-MB-231 cell line, showing higher nuclear fragmentation, ROS generation, and depletion of ΔΨm like apoptosis signs compared to nisin and with no cytotoxicity on normal cells. CONCLUSIONS The findings suggest that nisin delivery via PLGA nanoparticles can be used to treat cancer without significant effects on healthy cells.
Collapse
Affiliation(s)
- Tanweer Haider
- Department of Pharmaceutical Sciences, Dr. Harisingh Gour University, Sagar, Madhya Pradesh, India-470003
| | - Vikas Pandey
- Department of Pharmaceutical Sciences, Dr. Harisingh Gour University, Sagar, Madhya Pradesh, India-470003
| | - Chittaranjan Behera
- Formulation & Drug Delivery Division CSIR- Indian Institute of Integrative Medicine, Canal Road, Jammu, India-180001
| | - Pradeep Kumar
- Department of Pharmacy and Pharmacology, School of Therapeutic Sciences, Faculty of Health Sciences, University of the Witwatersrand, Johannesburg, South Africa
| | - Prem N Gupta
- Formulation & Drug Delivery Division CSIR- Indian Institute of Integrative Medicine, Canal Road, Jammu, India-180001
| | - Vandana Soni
- Department of Pharmaceutical Sciences, Dr. Harisingh Gour University, Sagar, Madhya Pradesh, India-470003
| |
Collapse
|
48
|
Zhou H, Huang R, Su T, Li B, Zhou H, Ren J, Li Z. A c-MWCNTs/AuNPs-based electrochemical cytosensor to evaluate the anticancer activity of pinoresinol from Cinnamomum camphora against HeLa cells. Bioelectrochemistry 2022; 146:108133. [DOI: 10.1016/j.bioelechem.2022.108133] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2021] [Revised: 02/23/2022] [Accepted: 04/07/2022] [Indexed: 11/26/2022]
|
49
|
Cyclic Peptides for the Treatment of Cancers: A Review. Molecules 2022; 27:molecules27144428. [PMID: 35889301 PMCID: PMC9317348 DOI: 10.3390/molecules27144428] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2022] [Revised: 07/03/2022] [Accepted: 07/07/2022] [Indexed: 02/04/2023] Open
Abstract
Cyclic peptides have been widely reported to have therapeutic abilities in the treatment of cancer. This has been proven through in vitro and in vivo studies against breast, lung, liver, colon, and prostate cancers, among others. The multitude of data available in the literature supports the potential of cyclic peptides as anticancer agents. This review summarizes the findings from previously reported studies and discusses the different cyclic peptide compounds, the sources, and their modes of action as anticancer agents. The prospects and future of cyclic peptides will also be described to give an overview on the direction of cyclic peptide development for clinical applications.
Collapse
|
50
|
Sadrekarimi H, Gardanova ZR, Bakhshesh M, Ebrahimzadeh F, Yaseri AF, Thangavelu L, Hasanpoor Z, Zadeh FA, Kahrizi MS. Emerging role of human microbiome in cancer development and response to therapy: special focus on intestinal microflora. Lab Invest 2022; 20:301. [PMID: 35794566 PMCID: PMC9258144 DOI: 10.1186/s12967-022-03492-7] [Citation(s) in RCA: 47] [Impact Index Per Article: 23.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2022] [Accepted: 06/20/2022] [Indexed: 12/12/2022]
Abstract
In recent years, there has been a greater emphasis on the impact of microbial populations inhabiting the gastrointestinal tract on human health and disease. According to the involvement of microbiota in modulating physiological processes (such as immune system development, vitamins synthesis, pathogen displacement, and nutrient uptake), any alteration in its composition and diversity (i.e., dysbiosis) has been linked to a variety of pathologies, including cancer. In this bidirectional relationship, colonization with various bacterial species is correlated with a reduced or elevated risk of certain cancers. Notably, the gut microflora could potentially play a direct or indirect role in tumor initiation and progression by inducing chronic inflammation and producing toxins and metabolites. Therefore, identifying the bacterial species involved and their mechanism of action could be beneficial in preventing the onset of tumors or controlling their advancement. Likewise, the microbial community affects anti-cancer approaches’ therapeutic potential and adverse effects (such as immunotherapy and chemotherapy). Hence, their efficiency should be evaluated in the context of the microbiome, underlining the importance of personalized medicine. In this review, we summarized the evidence revealing the microbiota's involvement in cancer and its mechanism. We also delineated how microbiota could predict colon carcinoma development or response to current treatments to improve clinical outcomes.
Collapse
|