1
|
Freuville L, Matthys C, Quinton L, Gillet JP. Venom-derived peptides for breaking through the glass ceiling of drug development. Front Chem 2024; 12:1465459. [PMID: 39398192 PMCID: PMC11468230 DOI: 10.3389/fchem.2024.1465459] [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: 07/16/2024] [Accepted: 09/04/2024] [Indexed: 10/15/2024] Open
Abstract
Venoms are complex mixtures produced by animals and consist of hundreds of components including small molecules, peptides, and enzymes selected for effectiveness and efficacy over millions of years of evolution. With the development of venomics, which combines genomics, transcriptomics, and proteomics to study animal venoms and their effects deeply, researchers have identified molecules that selectively and effectively act against membrane targets, such as ion channels and G protein-coupled receptors. Due to their remarkable physico-chemical properties, these molecules represent a credible source of new lead compounds. Today, not less than 11 approved venom-derived drugs are on the market. In this review, we aimed to highlight the advances in the use of venom peptides in the treatment of diseases such as neurological disorders, cardiovascular diseases, or cancer. We report on the origin and activity of the peptides already approved and provide a comprehensive overview of those still in development.
Collapse
Affiliation(s)
- Lou Freuville
- Laboratory of Mass Spectrometry, MolSys Research Unit, University of Liège, Liège, Belgium
| | - Chloé Matthys
- Laboratory of Molecular Cancer Biology, URPhyM, NARILIS, University of Namur, Namur, Belgium
| | - Loïc Quinton
- Laboratory of Mass Spectrometry, MolSys Research Unit, University of Liège, Liège, Belgium
| | - Jean-Pierre Gillet
- Laboratory of Molecular Cancer Biology, URPhyM, NARILIS, University of Namur, Namur, Belgium
| |
Collapse
|
2
|
Handy NB, Xu Y, Moon D, Sowizral JJ, Moon E, Ho M, Wilson BA. Hierarchical determinants in cytotoxic necrotizing factor (CNF) toxins driving Rho G-protein deamidation versus transglutamination. mBio 2024; 15:e0122124. [PMID: 38920360 PMCID: PMC11253639 DOI: 10.1128/mbio.01221-24] [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: 04/27/2024] [Accepted: 05/22/2024] [Indexed: 06/27/2024] Open
Abstract
The cytotoxic necrotizing factor (CNF) family of AB-type bacterial protein toxins catalyze two types of modification on their Rho GTPase substrates: deamidation and transglutamination. It has been established that E. coli CNF1 and its close homolog proteins catalyze primarily deamidation and Bordetella dermonecrotic toxin (DNT) catalyzes primarily transglutamination. The rapidly expanding microbial genome sequencing data have revealed that there are at least 13 full-length variants of CNF1 homologs. CNFx from E. coli strain GN02091 is the most distant from all other members of the CNF family with 50%-55% sequence identity at the protein level and 0.45-0.52 nucleotide substitutions per site at the DNA level. CNFx modifies RhoA, Rac1, and Cdc42, and like CNF1, activates downstream SRE-dependent mitogenic signaling pathways in human HEK293T cells, but at a 1,000-fold higher EC50 value. Unlike other previously characterized CNF toxins, CNFx modifies Rho proteins primarily through transglutamination, as evidenced by gel-shift assay and confirmed by MALDI mass spectral analysis, when coexpressed with Rho-protein substrates in E. coli BL21 cells or through direct treatment of HEK293T cells. A comparison of CNF1 and CNFx sequences identified two critical active-site residues corresponding to positions 832 and 862 in CNF1. Reciprocal site-specific mutations at these residues in each toxin revealed hierarchical rules that define the preference for deamidase versus a transglutaminase activity in CNFs. An additional unique Cys residue at the C-terminus of CNFx was also discovered to be critical for retarding cargo delivery.IMPORTANCECytotoxic necrotizing factor (CNF) toxins not only play important virulence roles in pathogenic E. coli and other bacterial pathogens, but CNF-like genes have also been found in an expanding number of genomes from clinical isolates. Harnessing the power of evolutionary relationships among the CNF toxins enabled the deciphering of the hierarchical active-site determinants that define whether they modify their Rho GTPase substrates through deamidation or transglutamination. With our finding that a distant CNF variant (CNFx) unlike other known CNFs predominantly transglutaminates its Rho GTPase substrates, the paradigm of "CNFs deamidate and DNTs transglutaminate" could finally be attributed to two critical amino acid residues within the active site other than the previously identified catalytic Cys-His dyad residues. The significance of our approach and research findings is that they can be applied to deciphering enzyme reaction determinants and substrate specificities for other bacterial proteins in the development of precision therapeutic strategies.
Collapse
Affiliation(s)
- Nicholas B. Handy
- Department of Microbiology, School of Molecular and Cellular Biology, University of Illinois at Urbana-Champaign, Urbana, Illinois, USA
| | - Yiting Xu
- Department of Microbiology, School of Molecular and Cellular Biology, University of Illinois at Urbana-Champaign, Urbana, Illinois, USA
| | - Damee Moon
- Department of Microbiology, School of Molecular and Cellular Biology, University of Illinois at Urbana-Champaign, Urbana, Illinois, USA
| | - Jacob J. Sowizral
- Department of Microbiology, School of Molecular and Cellular Biology, University of Illinois at Urbana-Champaign, Urbana, Illinois, USA
| | - Eric Moon
- Department of Microbiology, School of Molecular and Cellular Biology, University of Illinois at Urbana-Champaign, Urbana, Illinois, USA
| | - Mengfei Ho
- Department of Microbiology, School of Molecular and Cellular Biology, University of Illinois at Urbana-Champaign, Urbana, Illinois, USA
| | - Brenda A. Wilson
- Department of Microbiology, School of Molecular and Cellular Biology, University of Illinois at Urbana-Champaign, Urbana, Illinois, USA
| |
Collapse
|
3
|
Zhang R, Li X, Zhang S. The Role of Bacteria in Central Nervous System Tumors: Opportunities and Challenges. Microorganisms 2024; 12:1053. [PMID: 38930435 PMCID: PMC11205425 DOI: 10.3390/microorganisms12061053] [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: 05/05/2024] [Revised: 05/19/2024] [Accepted: 05/20/2024] [Indexed: 06/28/2024] Open
Abstract
Tumors of the central nervous system (CNS) are severe and refractory diseases with poor prognosis, especially for patients with malignant glioblastoma and brain metastases. Currently, numerous studies have explored the potential role of bacteria and intestinal flora in tumor development and treatment. Bacteria can penetrate the blood-brain barrier (BBB), targeting the hypoxic microenvironment at the core of tumors, thereby eliminating tumors and activating both the innate and adaptive immune responses, rendering them promising therapeutic agents for CNS tumors. In addition, engineered bacteria and derivatives, such as bacterial membrane proteins and bacterial spores, can also be used as good candidate carriers for targeted drug delivery. Moreover, the intestinal flora can regulate CNS tumor metabolism and influence the immune microenvironment through the "gut-brain axis". Therefore, bacterial anti-tumor therapy, engineered bacterial targeted drug delivery, and intervention of the intestinal flora provide therapeutic modalities for the treatment of CNS tumors. In this paper, we performed a comprehensive review of the mechanisms and therapeutic practices of bacterial therapy for CNS tumors and discussed potential future research directions in this field.
Collapse
Affiliation(s)
| | | | - Si Zhang
- Department of Neurosurgery, West China Hospital, Sichuan University, Chengdu 610041, China; (R.Z.); (X.L.)
| |
Collapse
|
4
|
Schmidt G. Some Examples of Bacterial Toxins as Tools. Toxins (Basel) 2024; 16:202. [PMID: 38787054 PMCID: PMC11125981 DOI: 10.3390/toxins16050202] [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: 03/05/2024] [Revised: 04/18/2024] [Accepted: 04/20/2024] [Indexed: 05/25/2024] Open
Abstract
Pathogenic bacteria produce diverse protein toxins to disturb the host's defenses. This includes the opening of epithelial barriers to establish bacterial growth in deeper tissues of the host and to modulate immune cell functions. To achieve this, many toxins share the ability to enter mammalian cells, where they catalyze the modification of cellular proteins. The enzymatic activity is diverse and ranges from ribosyl- or glycosyl-transferase activity, the deamidation of proteins, and adenylate-cyclase activity to proteolytic cleavage. Protein toxins are highly active enzymes often with tight specificity for an intracellular protein or a protein family coupled with the intrinsic capability of entering mammalian cells. A broad understanding of their molecular mechanisms established bacterial toxins as powerful tools for cell biology. Both the enzymatic part and the pore-forming/protein transport capacity are currently used as tools engineered to study signaling pathways or to transport cargo like labeled compounds, nucleic acids, peptides, or proteins directly into the cytosol. Using several representative examples, this review is intended to provide a short overview of the state of the art in the use of bacterial toxins or parts thereof as tools.
Collapse
Affiliation(s)
- Gudula Schmidt
- Institute of Experimental and Clinical Pharmacology and Toxicology, University of Freiburg, Albertstr. 25, 79104 Freiburg, Germany
| |
Collapse
|
5
|
Xia Z, He D, Wu Y, Kwok HF, Cao Z. Scorpion venom peptides: Molecular diversity, structural characteristics, and therapeutic use from channelopathies to viral infections and cancers. Pharmacol Res 2023; 197:106978. [PMID: 37923027 DOI: 10.1016/j.phrs.2023.106978] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/23/2023] [Revised: 10/23/2023] [Accepted: 10/27/2023] [Indexed: 11/07/2023]
Abstract
Animal venom is an important evolutionary innovation in nature. As one of the most representative animal venoms, scorpion venom contains an extremely diverse set of bioactive peptides. Scorpion venom peptides not only are 'poisons' that immobilize, paralyze, kill, or dissolve preys but also become important candidates for drug development and design. Here, the review focuses on the molecular diversity of scorpion venom peptides, their typical structural characteristics, and their multiple therapeutic or pharmaceutical applications in channelopathies, viral infections and cancers. Especially, the group of scorpion toxin TRPTx targeting transient receptor potential (TRP) channels is systematically summarized and worthy of attention because TRP channels play a crucial role in the regulation of homeostasis and the occurrence of diseases in human. We also further establish the potential relationship between the molecular characteristics and functional applications of scorpion venom peptides to provide a research basis for modern drug development and clinical utilization of scorpion venom resources.
Collapse
Affiliation(s)
- Zhiqiang Xia
- School of Biological and Food Processing Engineering, Huanghuai University, Zhumadian, China
| | - Dangui He
- State Key Laboratory of Virology, College of Life Sciences, Shenzhen Research Institute, Wuhan University, Wuhan, China; Department of Biomedical Sciences, Faculty of Health Sciences, University of Macau, Avenida de Universidade, Taipa, Macao; Cancer Centre, Faculty of Health Sciences, University of Macau, Avenida de Universidade, Taipa, Macao
| | - Yingliang Wu
- State Key Laboratory of Virology, College of Life Sciences, Shenzhen Research Institute, Wuhan University, Wuhan, China
| | - Hang Fai Kwok
- Department of Biomedical Sciences, Faculty of Health Sciences, University of Macau, Avenida de Universidade, Taipa, Macao; Cancer Centre, Faculty of Health Sciences, University of Macau, Avenida de Universidade, Taipa, Macao; MoE Frontiers Science Center for Precision Oncology, University of Macau, Avenida de Universidade, Taipa, Macao.
| | - Zhijian Cao
- State Key Laboratory of Virology, College of Life Sciences, Shenzhen Research Institute, Wuhan University, Wuhan, China; Bio-drug Research Center, Wuhan University, Wuhan, China.
| |
Collapse
|
6
|
Zhou Y, Zhang Y, Tian J, Miao Z, Lv S, Zhao X. A Meaningful Strategy for Glioma Diagnosis via Independent Determination of hsa_circ_0004214. Brain Sci 2023; 13:brainsci13020193. [PMID: 36831736 PMCID: PMC9954075 DOI: 10.3390/brainsci13020193] [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: 01/06/2023] [Accepted: 01/16/2023] [Indexed: 01/26/2023] Open
Abstract
Glioma is one of the most common primary tumors in the central nervous system. Circular RNAs (circRNAs) may serve as novel biomarkers of various cancers. The purpose of this study is to reveal the diagnostic value of hsa_circ_0004214 for glioma and to predict its molecular interaction network. The expression of hsa_circ_0004214 was evaluated by RT-qPCR. The vector and siRNAs changed the expression of hsa_circ_0004214 to judge its influence on the migration degree of glioma cells. hsa_circ_0004214 can be stably expressed at a high level in high-grade glioma tissue (WHO III/IV). The area under the ROC curve of hsa_circ_0000745 in glioma tissue was 0.88, suggesting good diagnostic value. While used to distinguish high-grade glioma, AUC value can be increased to 0.931. The multi-factor correlation analysis found that the expression of hsa_circ_0004214 was correlated with GFAP (+) and Ki67 (+) in immunohistochemistry. In addition, the migration capacity of U87 was enhanced by overexpression of hsa_circ_0004214. Through miRNA microarray analysis and database screening, we finally identified 4 miRNAs and 9 RBPs that were most likely to interact with hsa_circ_0004214 and regulate the biological functions of glioma. Hsa_circ_0004 214 plays an important role in glioma, its expression level is a promising diagnostic marker for this malignancy.
Collapse
Affiliation(s)
- Yinan Zhou
- Department of Neurosurgery, Medical School of Nantong University, Nantong University, Nantong 226019, China
- Wuxi No. 2 People’s Hospital, Affiliated Wuxi Clinical College of Nantong University, Wuxi 214002, China
| | - Yating Zhang
- Department of Neurosurgery, Medical School of Nantong University, Nantong University, Nantong 226019, China
- Wuxi No. 2 People’s Hospital, Affiliated Wuxi Clinical College of Nantong University, Wuxi 214002, China
| | - Jiajia Tian
- Wuxi Clinical Medical College of Nanjing Medical University, Nanjing Medical University, Wuxi 214002, China
| | - Zengli Miao
- Department of Neurosurgery, Medical School of Nantong University, Nantong University, Nantong 226019, China
- Wuxi No. 2 People’s Hospital, Affiliated Wuxi Clinical College of Nantong University, Wuxi 214002, China
| | - Shangrui Lv
- Wuxi Clinical Medical College of Nanjing Medical University, Nanjing Medical University, Wuxi 214002, China
| | - Xudong Zhao
- Department of Neurosurgery, Medical School of Nantong University, Nantong University, Nantong 226019, China
- Wuxi No. 2 People’s Hospital, Affiliated Wuxi Clinical College of Nantong University, Wuxi 214002, China
- Department of Neurosurgery, Wuxi No. 2 People’s Hospital, Wuxi 214002, China
- Correspondence:
| |
Collapse
|
7
|
Meng Y, Sun J, Zhang G, Yu T, Piao H. Bacteria associated with glioma: a next wave in cancer treatment. Front Cell Infect Microbiol 2023; 13:1164654. [PMID: 37201117 PMCID: PMC10185885 DOI: 10.3389/fcimb.2023.1164654] [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: 02/13/2023] [Accepted: 04/21/2023] [Indexed: 05/20/2023] Open
Abstract
Malignant gliomas occur more often in adults and may affect any part of the central nervous system (CNS). Although their results could be better, surgical excision, postoperative radiation and chemotherapy, and electric field therapy are today's mainstays of glioma care. However, bacteria can also exert anti-tumor effects via mechanisms such as immune regulation and bacterial toxins to promote apoptosis, inhibit angiogenesis, and rely on their natural characteristics to target the tumor microenvironment of hypoxia, low pH, high permeability, and immunosuppression. Tumor-targeted bacteria expressing anticancer medications will go to the cancer site, colonize the tumor, and then produce the therapeutic chemicals that kill the cancer cells. Targeting bacteria in cancer treatment has promising prospects. Rapid advances have been made in the study of bacterial treatment of tumors, including using bacterial outer membrane vesicles to load chemotherapy drugs or combine with nanomaterials to fight tumors, as well as the emergence of bacteria combined with chemotherapy, radiotherapy, and photothermal/photodynamic therapy. In this study, we look back at the previous years of research on bacteria-mediated glioma treatment and move forward to where we think it is headed.
Collapse
Affiliation(s)
- Yiming Meng
- Department of Central Laboratory, Cancer Hospital of Dalian University of Technology, Liaoning Cancer Hospital & Institute. No. 44, Shenyang, China
- *Correspondence: Yiming Meng, ; Tao Yu, ; Haozhe Piao,
| | - Jing Sun
- Department of Biobank, Cancer Hospital of Dalian University of Technology, Liaoning Cancer Hospital & Institute. No. 44, Shenyang, China
| | - Guirong Zhang
- Department of Central Laboratory, Cancer Hospital of Dalian University of Technology, Liaoning Cancer Hospital & Institute. No. 44, Shenyang, China
| | - Tao Yu
- Department of Medical Imaging, Cancer Hospital of Dalian University of Technology, Liaoning Cancer Hospital & Institute. No. 44, Shenyang, China
- *Correspondence: Yiming Meng, ; Tao Yu, ; Haozhe Piao,
| | - Haozhe Piao
- Department of Central Laboratory, Cancer Hospital of Dalian University of Technology, Liaoning Cancer Hospital & Institute. No. 44, Shenyang, China
- Department of Neurosurgery, Cancer Hospital of Dalian University of Technology, Liaoning Cancer Hospital & Institute. No. 44, Shenyang, China
- *Correspondence: Yiming Meng, ; Tao Yu, ; Haozhe Piao,
| |
Collapse
|
8
|
Chat H, Dalmasso G, Godfraind C, Bonnin V, Beyrouthy R, Bonnet M, Barnich N, Mettouchi A, Lemichez E, Bonnet R, Delmas J. Cytotoxic necrotizing factor 1 hinders colon tumorigenesis induced by colibactin-producing Escherichia coli in ApcMin/+ mice. Gut Microbes 2023; 15:2229569. [PMID: 37417545 PMCID: PMC10332217 DOI: 10.1080/19490976.2023.2229569] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/24/2022] [Accepted: 06/21/2023] [Indexed: 07/08/2023] Open
Abstract
Colorectal cancer (CRC) patients are frequently colonized by colibactin-producing Escherichia coli (CoPEC) (>40%), which enhances tumorigenesis in mouse models of CRC. We observed that 50% of CoPEC also contains the cnf1 gene, which encodes cytotoxic necrotizing factor-1 (CNF1), an enhancer of the eukaryotic cell cycle. The impact of its co-occurrence with colibactin (Clb) has not yet been investigated. We evaluated the impact of CNF1 on colorectal tumorigenesis using human colonic epithelial HT-29 cells and CRC-susceptible ApcMin/+ mice inoculated with the CoPEC 21F8 clinical strain (Clb+Cnf+) or 21F8 isogenic mutants (Clb+Cnf-, Clb-Cnf+ and Clb-Cnf-). Infection with the Clb+Cnf- strain induced higher levels of inflammatory cytokines and senescence markers both in vitro and in vivo compared to those induced by infection with the Clb+Cnf+ strain. In contrast, the Clb+Cnf- and Clb+Cnf+ strains generated similar levels of DNA damage in HT-29 cells and in colonic murine tissues. Furthermore, the ApcMin/+ mice inoculated with the Clb+Cnf- strain developed significantly more tumors than the mice inoculated with the Clb+Cnf+ strain or the isogenic mutants, and the composition of their microbiota was changed. Finally, rectal administration of the CNF1 protein in ApcMin/+ mice inoculated with the Clb+Cnf- strain significantly decreased tumorigenesis and inflammation. Overall, this study provides evidence that CNF1 decreases the carcinogenic effects of CoPEC in ApcMin/+ mice by decreasing CoPEC-induced cellular senescence and inflammation.
Collapse
Affiliation(s)
- Héloïse Chat
- Centre de Recherche en Nutrition Humaine Auvergne, University Clermont Auvergne, Inserm U1071, INRAE USC 1382, Microbes, Intestin, Inflammation et Susceptibilité de l’Hôte (M2iSH), Clermont-Ferrand, France
| | - Guillaume Dalmasso
- Centre de Recherche en Nutrition Humaine Auvergne, University Clermont Auvergne, Inserm U1071, INRAE USC 1382, Microbes, Intestin, Inflammation et Susceptibilité de l’Hôte (M2iSH), Clermont-Ferrand, France
| | - Catherine Godfraind
- Centre de Recherche en Nutrition Humaine Auvergne, University Clermont Auvergne, Inserm U1071, INRAE USC 1382, Microbes, Intestin, Inflammation et Susceptibilité de l’Hôte (M2iSH), Clermont-Ferrand, France
- Neuropathology Unit, University Hospital of Clermont-Ferrand, Clermont-Ferrand, France
| | - Virginie Bonnin
- Centre de Recherche en Nutrition Humaine Auvergne, University Clermont Auvergne, Inserm U1071, INRAE USC 1382, Microbes, Intestin, Inflammation et Susceptibilité de l’Hôte (M2iSH), Clermont-Ferrand, France
| | - Racha Beyrouthy
- Centre de Recherche en Nutrition Humaine Auvergne, University Clermont Auvergne, Inserm U1071, INRAE USC 1382, Microbes, Intestin, Inflammation et Susceptibilité de l’Hôte (M2iSH), Clermont-Ferrand, France
| | - Mathilde Bonnet
- Centre de Recherche en Nutrition Humaine Auvergne, University Clermont Auvergne, Inserm U1071, INRAE USC 1382, Microbes, Intestin, Inflammation et Susceptibilité de l’Hôte (M2iSH), Clermont-Ferrand, France
- Institut Universitaire de Technologie, University Clermont Auvergne, Clermont-Ferrand, France
| | - Nicolas Barnich
- Centre de Recherche en Nutrition Humaine Auvergne, University Clermont Auvergne, Inserm U1071, INRAE USC 1382, Microbes, Intestin, Inflammation et Susceptibilité de l’Hôte (M2iSH), Clermont-Ferrand, France
- Institut Universitaire de Technologie, University Clermont Auvergne, Clermont-Ferrand, France
| | - Amel Mettouchi
- Institut Pasteur, University of Paris, CNRS UMR2001, Paris, France
| | | | - Richard Bonnet
- Centre de Recherche en Nutrition Humaine Auvergne, University Clermont Auvergne, Inserm U1071, INRAE USC 1382, Microbes, Intestin, Inflammation et Susceptibilité de l’Hôte (M2iSH), Clermont-Ferrand, France
- Department of Bacteriology, University Hospital of Clermont-Ferrand, Clermont-Ferrand, France
| | - Julien Delmas
- Centre de Recherche en Nutrition Humaine Auvergne, University Clermont Auvergne, Inserm U1071, INRAE USC 1382, Microbes, Intestin, Inflammation et Susceptibilité de l’Hôte (M2iSH), Clermont-Ferrand, France
- Department of Bacteriology, University Hospital of Clermont-Ferrand, Clermont-Ferrand, France
| |
Collapse
|
9
|
Exploiting the Endogenous Ubiquitin Proteasome System in Targeted Cancer Treatment. Cancers (Basel) 2022; 15:cancers15010256. [PMID: 36612252 PMCID: PMC9818074 DOI: 10.3390/cancers15010256] [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: 11/13/2022] [Revised: 12/15/2022] [Accepted: 12/26/2022] [Indexed: 01/03/2023] Open
Abstract
To overcome the lack of specificity of cancer therapeutics and thus create a more potent and effective treatment, we developed a novel chimeric protein, IL2-Smurf2. Here, we describe the production of this chimeric IL2-Smurf2 protein and its variants, with inactive or over-active killing components. Using Western blots, we demonstrated the chimeric protein's ability to specifically enter target cells alone. After entering the cells, the protein showed biological activity, causing cell death that was not seen with an inactive variant, and that was shown to be apoptotic. The chimeric protein also proved to be active as an E3 ligase, as demonstrated by testing total ubiquitination levels along with targeted ubiquitination for degradation. Finally, we tested IL2-Smurf2 and its variants in an in vivo mouse model of leukemia and demonstrated its potential as a drug for the targeted treatment of cancer cells. In the course of this work, we established for the first time the feasibility of the use of Smurf2 as a killing component in chimeric targeting proteins. Utilizing the IL2 cytokine to target cells overexpressing IL-2R and Smurf2 to cause protein degradation, we were able to produce a chimeric protein with dual functionality which causes targeted cell death.
Collapse
|
10
|
Investigational Microbiological Therapy for Glioma. Cancers (Basel) 2022; 14:cancers14235977. [PMID: 36497459 PMCID: PMC9736089 DOI: 10.3390/cancers14235977] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2022] [Revised: 11/27/2022] [Accepted: 11/29/2022] [Indexed: 12/07/2022] Open
Abstract
Glioma is the most common primary malignancy of the central nervous system (CNS), and 50% of patients present with glioblastoma (GBM), which is the most aggressive type. Currently, the most popular therapies are progressive chemotherapy and treatment with temozolomide (TMZ), but the median survival of glioma patients is still low as a result of the emergence of drug resistance, so we urgently need to find new therapies. A growing number of studies have shown that the diversity, bioactivity, and manipulability of microorganisms make microbial therapy a promising approach for cancer treatment. However, the many studies on the research progress of microorganisms and their derivatives in the development and treatment of glioma are scattered, and nobody has yet provided a comprehensive summary of them. Therefore, in this paper, we review the research progress of microorganisms and their derivatives in the development and treatment of glioma and conclude that it is possible to treat glioma by exogenous microbial therapies and targeting the gut-brain axis. In this article, we discuss the prospects and pressing issues relating to these therapies with the aim of providing new ideas for the treatment of glioma.
Collapse
|
11
|
Markelova NN, Semenova EF, Sineva ON, Sadykova VS. The Role of Cyclomodulins and Some Microbial Metabolites in Bacterial Microecology and Macroorganism Carcinogenesis. Int J Mol Sci 2022; 23:ijms231911706. [PMID: 36233008 PMCID: PMC9570213 DOI: 10.3390/ijms231911706] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2022] [Revised: 09/24/2022] [Accepted: 09/26/2022] [Indexed: 11/16/2022] Open
Abstract
A number of bacteria that colonize the human body produce toxins and effectors that cause changes in the eukaryotic cell cycle—cyclomodulins and low-molecular-weight compounds such as butyrate, lactic acid, and secondary bile acids. Cyclomodulins and metabolites are necessary for bacteria as adaptation factors—which are influenced by direct selection—to the ecological niches of the host. In the process of establishing two-way communication with the macroorganism, these compounds cause limited damage to the host, despite their ability to disrupt key processes in eukaryotic cells, which can lead to pathological changes. Possible negative consequences of cyclomodulin and metabolite actions include their potential role in carcinogenesis, in particular, with the ability to cause DNA damage, increase genome instability, and interfere with cancer-associated regulatory pathways. In this review, we aim to examine cyclomodulins and bacterial metabolites as important factors in bacterial survival and interaction with the host organism to show their heterogeneous effect on oncogenesis depending on the surrounding microenvironment, pathological conditions, and host genetic background.
Collapse
Affiliation(s)
- Natalia N. Markelova
- Gause Institute of New Antibiotics, ul. Bolshaya Pirogovskaya, 11, 119021 Moscow, Russia
- Correspondence: (N.N.M.); (V.S.S.)
| | - Elena F. Semenova
- Institute of Biochemical Technology, Ecology and Pharmacy, V.I. Vernadsky Crimean Federal University, 295007 Simferopol, Russia
| | - Olga N. Sineva
- Gause Institute of New Antibiotics, ul. Bolshaya Pirogovskaya, 11, 119021 Moscow, Russia
| | - Vera S. Sadykova
- Gause Institute of New Antibiotics, ul. Bolshaya Pirogovskaya, 11, 119021 Moscow, Russia
- Correspondence: (N.N.M.); (V.S.S.)
| |
Collapse
|
12
|
Hua T, Shi H, Zhu M, Chen C, Su Y, Wen S, Zhang X, Chen J, Huang Q, Wang H. Glioma‑neuronal interactions in tumor progression: Mechanism, therapeutic strategies and perspectives (Review). Int J Oncol 2022; 61:104. [PMID: 35856439 PMCID: PMC9339490 DOI: 10.3892/ijo.2022.5394] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2022] [Accepted: 06/30/2022] [Indexed: 11/06/2022] Open
Abstract
An increasing body of evidence has become available to reveal the synaptic and functional integration of glioma into the brain network, facilitating tumor progression. The novel discovery of glioma-neuronal interactions has fundamentally challenged our understanding of this refractory disease. The present review aimed to provide an overview of how the neuronal activities function through synapses, neurotransmitters, ion channels, gap junctions, tumor microtubes and neuronal molecules to establish communications with glioma, as well as a simplified explanation of the reciprocal effects of crosstalk on neuronal pathophysiology. In addition, the current state of therapeutic avenues targeting critical factors involved in glioma-euronal interactions is discussed and an overview of clinical trial data for further investigation is provided. Finally, newly emerging technologies, including immunomodulation, a neural stem cell-based delivery system, optogenetics techniques and co-culture of neuron organoids and glioma, are proposed, which may pave a way towards gaining deeper insight into both the mechanisms associated with neuron- and glioma-communicating networks and the development of therapeutic strategies to target this currently lethal brain tumor.
Collapse
Affiliation(s)
- Tianzhen Hua
- Department of Neurosurgery, Changhai Hospital, Naval Medical University, Shanghai 200433, P.R. China
| | - Huanxiao Shi
- Department of Neurosurgery, Changhai Hospital, Naval Medical University, Shanghai 200433, P.R. China
| | - Mengmei Zhu
- Department of Neurosurgery, Changhai Hospital, Naval Medical University, Shanghai 200433, P.R. China
| | - Chao Chen
- Department of Neurosurgery, Changhai Hospital, Naval Medical University, Shanghai 200433, P.R. China
| | - Yandong Su
- Department of Neurosurgery, Changhai Hospital, Naval Medical University, Shanghai 200433, P.R. China
| | - Shengjia Wen
- Department of Neurosurgery, Changhai Hospital, Naval Medical University, Shanghai 200433, P.R. China
| | - Xu Zhang
- Department of Neurosurgery, Changhai Hospital, Naval Medical University, Shanghai 200433, P.R. China
| | - Juxiang Chen
- Department of Neurosurgery, Changhai Hospital, Naval Medical University, Shanghai 200433, P.R. China
| | - Qilin Huang
- Department of Neurosurgery, General Hospital of Central Theater Command of Chinese People's Liberation Army, Wuhan, Hubei 430070, P.R. China
| | - Hongxiang Wang
- Department of Neurosurgery, Changhai Hospital, Naval Medical University, Shanghai 200433, P.R. China
| |
Collapse
|
13
|
Degl’Innocenti E, Poloni TE, Medici V, Recupero L, Dell’Amico C, Vannini E, Borello U, Mazzanti CM, Onorati M, Dell’Anno MT. Centrin 2: A Novel Marker of Mature and Neoplastic Human Astrocytes. Front Cell Neurosci 2022; 16:858347. [PMID: 35573835 PMCID: PMC9100563 DOI: 10.3389/fncel.2022.858347] [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: 01/19/2022] [Accepted: 03/17/2022] [Indexed: 11/13/2022] Open
Abstract
As microtubule-organizing centers (MTOCs), centrosomes play a pivotal role in cell division, neurodevelopment and neuronal maturation. Among centrosomal proteins, centrin-2 (CETN2) also contributes to DNA repair mechanisms which are fundamental to prevent genomic instability during neural stem cell pool expansion. Nevertheless, the expression profile of CETN2 in human neural stem cells and their progeny is currently unknown. To address this question, we interrogated a platform of human neuroepithelial stem (NES) cells derived from post mortem developing brain or established from pluripotent cells and demonstrated that while CETN2 retains its centrosomal location in proliferating NES cells, its expression pattern changes upon differentiation. In particular, we found that CETN2 is selectively expressed in mature astrocytes with a broad cytoplasmic distribution. We then extended our findings on human autoptic nervous tissue samples. We investigated CETN2 distribution in diverse anatomical areas along the rostro-caudal neuraxis and pointed out a peculiar topography of CETN2-labeled astrocytes in humans which was not appreciable in murine tissues, where CETN2 was mostly confined to ependymal cells. As a prototypical condition with glial overproliferation, we also explored CETN2 expression in glioblastoma multiforme (GBM), reporting a focal concentration of CETN2 in neoplastic astrocytes. This study expands CETN2 localization beyond centrosomes and reveals a unique expression pattern that makes it eligible as a novel astrocytic molecular marker, thus opening new roads to glial biology and human neural conditions.
Collapse
Affiliation(s)
- Elisa Degl’Innocenti
- Fondazione Pisana per la Scienza ONLUS, San Giuliano Terme, Italy
- Department of Translational Research and of New Technologies in Medicine and Surgery, University of Pisa, Pisa, Italy
| | - Tino Emanuele Poloni
- Department of Neurology and Neuropathology, Golgi-Cenci Foundation and ASP Golgi-Redaelli, Abbiategrasso, Italy
| | - Valentina Medici
- Department of Neurology and Neuropathology, Golgi-Cenci Foundation and ASP Golgi-Redaelli, Abbiategrasso, Italy
| | - Luca Recupero
- Fondazione Pisana per la Scienza ONLUS, San Giuliano Terme, Italy
| | - Claudia Dell’Amico
- Unit of Cell and Developmental Biology, Department of Biology, University of Pisa, Pisa, Italy
| | | | - Ugo Borello
- Unit of Cell and Developmental Biology, Department of Biology, University of Pisa, Pisa, Italy
| | | | - Marco Onorati
- Unit of Cell and Developmental Biology, Department of Biology, University of Pisa, Pisa, Italy
| | - Maria Teresa Dell’Anno
- Fondazione Pisana per la Scienza ONLUS, San Giuliano Terme, Italy
- *Correspondence: Maria Teresa Dell’Anno,
| |
Collapse
|