Next Article in Journal
Genomic Analysis of Romanian Lycium Genotypes: Exploring BODYGUARD Genes for Stress Resistance Breeding
Previous Article in Journal
The Potential Use of Vitamin D3 and Phytochemicals for Their Anti-Ageing Effects
Previous Article in Special Issue
AB Toxins as High-Affinity Ligands for Cell Targeting in Cancer Therapy
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Editorial

Special Issue “Bacterial Toxins and Cancer”

Department of Cardiovascular, Endocrine-Metabolic Diseases and Aging, Istituto Superiore di Sanità, 00161 Rome, Italy
*
Author to whom correspondence should be addressed.
Int. J. Mol. Sci. 2024, 25(4), 2128; https://doi.org/10.3390/ijms25042128
Submission received: 24 January 2024 / Accepted: 5 February 2024 / Published: 9 February 2024
(This article belongs to the Special Issue Bacterial Toxins and Cancer)
Infection is a major contributor to the development of cancer, with more than 15% of new cancer diagnoses estimated to be caused by infection [1,2,3]. Eleven infectious biological agents are classified as “Group 1 carcinogens” in the Monographs of the International Agency for Research on Cancer. These include viruses, such as Epstein–Barr, hepatitis B and C viruses, human immunodeficiency virus type 1, the Helicobacter pylori bacterium, and helminths infections [4]. Bacteria are increasingly recognized in the field of oncology, and several studies have been conducted on the pro-carcinogenic potential of chronic bacterial infections in certain districts [5,6,7,8,9,10]. Virulence factors produced by pathogenic bacteria as protein toxins favor colonization and diffusion in the host, disturbing the physiological balance and causing a state of disease. In this context, some bacterial toxins have been studied in recent years because of their association with cancer initiation and progression [11,12,13,14,15,16,17,18]. These toxins generally induce changes in cells that are reminiscent of cancer cells and possess highly specific enzymatic activity on key molecular targets. In the rapidly evolving landscape of molecular biology studies, bacterial toxins are also being considered as novel potential therapeutics, particularly in the field of anti-cancer therapies [19,20,21,22,23].
The current Special Issue, entitled “Bacterial Toxins and Cancer”, collects insightful reviews and research articles on the association between bacterial toxins and cancer initiation and progression, as well as the ability of some bacterial toxins to serve as useful tools for molecular biology studies by manipulating key host cellular processes. Six recent research papers on different topics have been included, specifically three research articles [24,25,26] and three reviews [27,28,29], with over 30 different contributors, that explore recent advances in the field of bacterial toxins and cancer in both the aforementioned areas. In fact, four papers address the potential anti-cancer activity of toxins [24,25,26,27], and two focus on the possible involvement of these virulence factors in cancer initiation and progression [28,29].
This summary provides a concise overview of the valuable papers in this Special Issue, with the goal of increasing their impact and citation rates.
In the field of toxins with anti-cancer potential, Xu et al. [24] focus on two interesting therapeutic targets that are overexpressed in pancreatic cancer (PC) and associated with a poor prognosis: the human epidermal growth factor receptor 3 (HER3) and the epithelial cell adhesion molecule (EpCAM). The authors analyze the anti-tumor effect of Ec1-LoPE (a fusion protein consisting of the EpCAM targeting agent Ec1 fused to the deimmunized cytotoxic agent LoPE toxin from Pseudomonas aeruginosa) and MM-121 (Seribantumab, an antagonist that blocks the ligand-dependent activation of HER3), either as monotherapy or in combination, in an in vivo mouse model of PC. The hypothesis is that, by targeting two different molecular mechanisms with a combined treatment, the cytotoxic effect on cancer cells would be enhanced, off-target toxicities would be potentially reduced, and the development of drug resistance would be prevented. The results show a significant effect of the combined therapy on the outcome parameters, but not on the survival curve of the treated mice. The authors conclude that the combined therapy with Ec1-LoPE and MM-21 is feasible for the treatment of PC, one of the most aggressive tumors with increasing incidence and a high mortality rate [30,31]. However, further investigation and optimization of the pharmacokinetic parameters are required.
The paper by Jun et al. [25] addresses one of the major limitations of the immunotoxins (ITs) in cancer therapy, namely, the concomitant expression of tumor antigen target receptors on normal tissues, resulting in on-target/off-tumor toxicity. To increase the selectivity of ITs for tumors over normal tissues, the authors propose a strategy to customize the anti-EGFR affinity of a monobody-based IT bearing a fragment of P. aeruginosa exotoxin A (PE24), termed ER-PE24, to distinguish solid tumors, which commonly overexpress EGFR, from normal tissues.
The authors compare the anti-tumor efficacy and toxicity of a panel of EGFR-targeting monobody-based PE24 toxins with different EGFR affinities. They found that ER/21-PE24, which is an intermediate EGFR affinity, shows strong anti-tumor activity with low systemic toxicity. In vitro and in vivo, they demonstrate eight- and fourfold improved therapeutic indices, respectively, compared to ER/0.2-PE24, which has the highest EGFR affinity.
The authors emphasize the importance of adjusting the EGFR affinity of EGFR-targeted ITs based on EGFR expression levels on tumors to maximize the therapeutic window while minimizing the on-target/off-tumor adverse effects.
Codolo et al. [26] present their findings regarding melanoma reduction after a treatment based on a protein toxin from Helicobacter pylori in an in vivo model.
The H. pylori neutrophil-activating protein (HP-NAP) has immunomodulatory properties [32] and may have potential therapeutic applications in the treatment of metastatic breast cancer and neuroendocrine tumors. It has been shown that HP-NAP inhibits the growth of bladder cancer and enhances the anti-tumor activity of oncolytic viruses by activating cytotoxic Th1 responses [33,34,35].
The authors investigate the potential for HP-NAP to exert its anti-tumor effect by modulating the activity of innate immune cells. Analyzing the larval stage of zebrafish, before the maturation of adaptive immunity, the authors demonstrate that HP-NAP administration effectively inhibits tumor growth and metastasis, increasing overall animal survival. This effect is accompanied by a significant recruitment of macrophages with pro-inflammatory profiles at the tumor site.
Therefore, this research indicates that reprogramming tumor macrophages could be an innovative strategy to successfully counteract melanoma growth, even in the absence of the acquired/specific branch of the immune system. Additionally, the bacterial protein HP-NAP could become a new biological therapeutic agent for the treatment of metastatic melanomas [36,37], either alone or in combination with immune checkpoint blockers.
Marquez-Lopez and Fanarraga [27] propose a review article on the potential uses of bacterial AB toxins in cancer therapy [38]. The authors describe the structure and mechanisms of action of the diphtheria (Dtx) [39], anthrax (Atx) [40], shiga (Stx) [41], and cholera (Ctx) [42] toxins and discuss the potential development of innovative and precise applications in oncology based on engineered recombinant AB toxins.
AB toxins are toxic molecules that strongly bind to cell receptors, some of which are overexpressed by both tumor cells and neovascular endothelial cells in solid tumors. Due to their modular structure, AB toxins can be genetically engineered to develop high-affinity compounds that can recognize target cells and facilitate the translocation of various therapeutic payloads into the cell. Among the toxins being considered, Stx is widely used both as an imaging and contrast agent, and it is also used as an attractive antineoplastic targeting tool [43]. Atx is the second most utilized, while the applications of Dtx and Ctx in cancer therapy are not as advanced as those of other toxins. Although there are several critical drawbacks that need to be addressed before the widespread use of engineered recombinant AB toxins in oncology, the results reported in this review suggest the possibility of developing innovative and promising applications in cancer treatment.
Markelova et al. [28] review the potential malignant role of toxins in the onset and progression of carcinogenesis, with a focus on cyclomodulins and bacterial metabolites as important factors in bacterial survival and interactions with the host organism. Strong associations between bacterial pathogens and cancer incidence are well known. Many bacterial factors have been identified that mediate the malignant transformation of host cells, including toxins, cell surface components, and effector proteins [44]. These factors are capable of causing DNA damage, increasing genome instability, and interfering with cancer-associated regulatory pathways. Even commensal bacteria, which have co-evolved with their hosts and produce a number of compounds that support various physiological processes in the human body, are capable of overcoming protective host responses and exerting pathological effects when external and/or internal factors intervene to disrupt macroorganism–metabolite homeostasis [45]. In this way, they influence the onset or development of pathological conditions, including neoplasia.
The authors emphasize the role of cyclomodulins and other bacterial metabolites in the development of pathological effects, including the onset and progression of malignant transformation in the host organism.
Finally, the review proposed by Fettucciari et al. [29] focuses on the senescence induced by Clostridium difficile toxin B [46] as a pathological player in the onset of colorectal cancer (CRC). The B toxin is responsible for the major pathogenic effects of C. difficile infection (CDI) [47], and is associated with irritable bowel syndrome (IBS) and inflammatory bowel diseases (IBDs) [48]. In addition, the B toxin has the ability to induce and increase the burden of senescent cells [46,49]. The authors discuss how the induction and accumulation of senescent cells in IBS and/or IBD after CDI and its recurrences could be a potential factor in the development of CRC. The initial accumulation of senescent cells may exert an anti-tumor effect by inducing the clearance of pre-malignant cells by immune cells recruited by some components of the senescence-associated secretory phenotype (SASP). However, in conditions where senescent cells accumulate (such as chronic injury, deregulation of immune surveillance, aging, etc.), the same SASP factors may promote tumor growth and drive cells towards complete CRC transformation [50,51]. Finally, the authors propose the elimination of senescent cells and/or neutralization of SASP as a possible therapeutic approach for CRC.
In summary, the articles included in this collection cover important research areas in the field of bacterial toxins and cancer. The field of research is still in its infancy, but bacterial toxins have proven to be highly versatile, acting both as carcinogens and, when appropriately modified, as highly specific anti-cancer agents.

Author Contributions

Conceptualization, S.T. and A.F.; validation, S.T. and A.F.; writing—original draft preparation, F.C. and Z.M.; writing—review and editing, S.T., F.C., Z.M. and A.F.; supervision, A.F. All authors have read and agreed to the published version of the manuscript.

Conflicts of Interest

The authors declare no conflicts of interest.

References

  1. Plummer, M.; de Martel, C.; Vignat, J.; Ferlay, J.; Bray, F.; Franceschi, S. Global Burden of Cancers Attributable to Infections in 2012: A Synthetic Analysis. Lancet Glob. Health 2016, 4, e609–e616. [Google Scholar] [CrossRef] [PubMed]
  2. Jacqueline, C.; Tasiemski, A.; Sorci, G.; Ujvari, B.; Maachi, F.; Missé, D.; Renaud, F.; Ewald, P.; Thomas, F.; Roche, B. Infections and Cancer: The “Fifty Shades of Immunity” Hypothesis. BMC Cancer 2017, 17, 257. [Google Scholar] [CrossRef] [PubMed]
  3. Emanuele Liardo, R.L.; Borzì, A.M.; Spatola, C.; Martino, B.; Privitera, G.; Basile, F.; Biondi, A.; Vacante, M. Effects of Infections on the Pathogenesis of Cancer. Indian J. Med. Res. 2021, 153, 431–445. [Google Scholar] [CrossRef] [PubMed]
  4. IARC Monographs on the Evaluation of Carcinogenic Risks to Humans Biological Agents; International Agency for Research on Cancer: Lyon, France, 2012; Volume 100 B, ISBN 978-92-832-1319-2.
  5. Baima, G.; Minoli, M.; Michaud, D.S.; Aimetti, M.; Sanz, M.; Loos, B.G.; Romandini, M. Periodontitis and Risk of Cancer: Mechanistic Evidence. Periodontology 2000 2023. [Google Scholar] [CrossRef] [PubMed]
  6. He, J.; Li, H.; Jia, J.; Liu, Y.; Zhang, N.; Wang, R.; Qu, W.; Liu, Y.; Jia, L. Mechanisms by Which the Intestinal Microbiota Affects Gastrointestinal Tumours and Therapeutic Effects. Mol. Biomed. 2023, 4, 45. [Google Scholar] [CrossRef] [PubMed]
  7. Sommariva, M.; Le Noci, V.; Bianchi, F.; Camelliti, S.; Balsari, A.; Tagliabue, E.; Sfondrini, L. The Lung Microbiota: Role in Maintaining Pulmonary Immune Homeostasis and Its Implications in Cancer Development and Therapy. Cell. Mol. Life Sci. 2020, 77, 2739–2749. [Google Scholar] [CrossRef] [PubMed]
  8. McLean, A.E.B.; Kao, S.C.; Barnes, D.J.; Wong, K.K.H.; Scolyer, R.A.; Cooper, W.A.; Kohonen-Corish, M.R.J. The Emerging Role of the Lung Microbiome and Its Importance in Non-Small Cell Lung Cancer Diagnosis and Treatment. Lung Cancer 2022, 165, 124–132. [Google Scholar] [CrossRef] [PubMed]
  9. Ding, R.; Lian, S.B.; Tam, Y.C.; Oh, C.C. The Cutaneous Microbiome in Skin Cancer—A Systematic Review. JDDG J. Dtsch. Dermatol. Ges. 2024. [Google Scholar] [CrossRef]
  10. Chalif, J.; Wang, H.; Spakowicz, D.; Quick, A.; Arthur, E.K.; O’malley, D.; Chambers, L.M. The Microbiome and Gynecologic Cancer: Cellular Mechanisms and Clinical Applications. Int. J. Gynecol. Cancer 2023, 34, 317–327. [Google Scholar] [CrossRef]
  11. Khatun, S.; Appidi, T.; Rengan, A.K. The Role Played by Bacterial Infections in the Onset and Metastasis of Cancer. Curr. Res. Microb. Sci. 2021, 2, 100078. [Google Scholar] [CrossRef]
  12. Lai, Y.R.; Chang, Y.F.; Ma, J.; Chiu, C.H.; Kuo, M.L.; Lai, C.H. From DNA Damage to Cancer Progression: Potential Effects of Cytolethal Distending Toxin. Front. Immunol. 2021, 12, 760451. [Google Scholar] [CrossRef]
  13. El Tekle, G.; Garrett, W.S. Bacteria in Cancer Initiation, Promotion and Progression. Nat. Rev. Cancer 2023, 23, 600–618. [Google Scholar] [CrossRef] [PubMed]
  14. Pleguezuelos-Manzano, C.; Puschhof, J.; Rosendahl Huber, A.; van Hoeck, A.; Wood, H.M.; Nomburg, J.; Gurjao, C.; Manders, F.; Dalmasso, G.; Stege, P.B.; et al. Mutational Signature in Colorectal Cancer Caused by Genotoxic Pks + E. Coli. Nature 2020, 580, 269–273. [Google Scholar] [CrossRef] [PubMed]
  15. Dalmasso, G.; Cougnoux, A.; Delmas, J.; Darfeuille-Michaud, A.; Bonnet, R. The Bacterial Genotoxin Colibactin Promotes Colon Tumor Growth by Modifying the Tumor Microenvironment. Gut Microbes 2014, 5, 675–680. [Google Scholar] [CrossRef] [PubMed]
  16. Purcell, R.V.; Pearson, J.; Aitchison, A.; Dixon, L.; Frizelle, F.A.; Keenan, J.I. Colonization with Enterotoxigenic Bacteroides Fragilis Is Associated with Early-Stage Colorectal Neoplasia. PLoS ONE 2017, 12, e0171602. [Google Scholar] [CrossRef] [PubMed]
  17. Zamani, S.; Taslimi, R.; Sarabi, A.; Jasemi, S.; Sechi, L.A.; Feizabadi, M.M. Enterotoxigenic Bacteroides Fragilis: A Possible Etiological Candidate for Bacterially-Induced Colorectal Precancerous and Cancerous Lesions. Front. Cell. Infect. Microbiol. 2019, 9, 449. [Google Scholar] [CrossRef] [PubMed]
  18. Lu, R.; Wu, S.; Zhang, Y.G.; Xia, Y.; Zhou, Z.; Kato, I.; Dong, H.; Bissonnette, M.; Sun, J. Salmonella Protein AvrA Activates the STAT3 Signaling Pathway in Colon Cancer. Neoplasia 2016, 18, 307–316. [Google Scholar] [CrossRef] [PubMed]
  19. Vannini, E.; Maltese, F.; Olimpico, F.; Fabbri, A.; Costa, M.; Caleo, M.; Baroncelli, L. Progression of Motor Deficits in Glioma-Bearing Mice: Impact of CNF1 Therapy at Symptomatic Stages. Oncotarget 2017, 8, 23539–23550. [Google Scholar] [CrossRef]
  20. Zhao, C.C.; Yu, W.W.; Qi, Y.J.; Xu, L.F.; Wang, Z.R.; Qiang, Y.W.; Ma, F.; Ma, X.L. Quantitative Proteomic Analysis Reveals That Luks-PV Exerts Antitumor Activity by Regulating the Key Proteins and Metabolic Pathways in HepG2 Cells. Anti-Cancer Drugs 2020, 31, 223–230. [Google Scholar] [CrossRef]
  21. Sharma, P.C.; Sharma, D.; Sharma, A.; Bhagat, M.; Ola, M.; Thakur, V.K.; Bhardwaj, J.K.; Goyal, R.K. Recent Advances in Microbial Toxin-Related Strategies to Combat Cancer. Semin. Cancer Biol. 2022, 86, 753–768. [Google Scholar] [CrossRef]
  22. Gao, Z.; Mcclane, B.A. Use of Clostridium Perfringens Enterotoxin and the Enterotoxin Receptor-Binding Domain (C-CPE) for Cancer Treatment: Opportunities and Challenges. J. Toxicol. 2012, 2012, 981626. [Google Scholar] [CrossRef] [PubMed]
  23. Weerakkody, L.; Witharana, C. The Role of Bacterial Toxins and Spores in Cancer Therapy. Life Sci. 2019, 235, 116839. [Google Scholar] [CrossRef] [PubMed]
  24. Xu, T.; Schulga, A.; Konovalova, E.; Rinne, S.S.; Zhang, H.; Vorontsova, O.; Orlova, A.; Deyev, S.M.; Tolmachev, V.; Vorobyeva, A. Feasibility of Co-Targeting HER3 and EpCAM Using Seribantumab and DARPin–Toxin Fusion in a Pancreatic Cancer Xenograft Model. Int. J. Mol. Sci. 2023, 24, 2838. [Google Scholar] [CrossRef] [PubMed]
  25. Jun, S.Y.; Kim, D.S.; Kim, Y.S. Expanding the Therapeutic Window of EGFR-Targeted PE24 Immunotoxin for EGFR-Overexpressing Cancers by Tailoring the EGFR Binding Affinity. Int. J. Mol. Sci. 2022, 23, 15820. [Google Scholar] [CrossRef]
  26. Codolo, G.; Facchinello, N.; Papa, N.; Bertocco, A.; Coletta, S.; Benna, C.; Dall’olmo, L.; Mocellin, S.; Tiso, N.; de Bernard, M. Macrophage-Mediated Melanoma Reduction after HP-NAP Treatment in a Zebrafish Xenograft Model. Int. J. Mol. Sci. 2022, 23, 1644. [Google Scholar] [CrossRef] [PubMed]
  27. Márquez-López, A.; Fanarraga, M.L. AB Toxins as High-Affinity Ligands for Cell Targeting in Cancer Therapy. Int. J. Mol. Sci. 2023, 24, 11227. [Google Scholar] [CrossRef] [PubMed]
  28. Markelova, N.N.; Semenova, E.F.; Sineva, O.N.; Sadykova, V.S. The Role of Cyclomodulins and Some Microbial Metabolites in Bacterial Microecology and Macroorganism Carcinogenesis. Int. J. Mol. Sci. 2022, 23, 11706. [Google Scholar] [CrossRef]
  29. Fettucciari, K.; Fruganti, A.; Stracci, F.; Spaterna, A.; Marconi, P.; Bassotti, G. Clostridioides Difficile Toxin B Induced Senescence: A New Pathologic Player for Colorectal Cancer? Int. J. Mol. Sci. 2023, 24, 8155. [Google Scholar] [CrossRef]
  30. Lambert, A.; Schwarz, L.; Borbath, I.; Henry, A.; Van Laethem, J.L.; Malka, D.; Ducreux, M.; Conroy, T. An Update on Treatment Options for Pancreatic Adenocarcinoma. Ther. Adv. Med. Oncol. 2019, 11, 1758835919875568. [Google Scholar] [CrossRef]
  31. Ansari, D.; Tingstedt, B.; Andersson, B.; Holmquist, F.; Sturesson, C.; Williamsson, C.; Sasor, A.; Borg, D.; Bauden, M.; Andersson, R. Pancreatic Cancer: Yesterday, Today and Tomorrow. Future Oncol. 2016, 12, 1929–1946. [Google Scholar] [CrossRef]
  32. D’Elios, M.M.; Amedei, A.; Cappon, A.; Del Prete, G.; De Bernard, M. The Neutrophil-Activating Protein of Helicobacter Pylori (HP-NAP) as an Immune Modulating Agent. FEMS Immunol. Med. Microbiol. 2007, 50, 157–164. [Google Scholar] [CrossRef] [PubMed]
  33. Ramachandran, M.; Yu, D.; Wanders, A.; Essand, M.; Eriksson, F. An Infection-Enhanced Oncolytic Adenovirus Secreting H. Pylori Neutrophil-Activating Protein with Therapeutic Effects on Neuroendocrine Tumors. Mol. Ther. 2013, 21, 2008–2018. [Google Scholar] [CrossRef] [PubMed]
  34. Amedei, A.; Cappon, A.; Codolo, G.; Cabrelle, A.; Polenghi, A.; Benagiano, M.; Tasca, E.; Azzurri, A.; Milco D’elios, M.; Del Prete, G.; et al. The Neutrophil-Activating Protein of Helicobacter Pylori Promotes Th1 Immune Responses. J. Clin. Investig. 2006, 116, 1092–1101. [Google Scholar] [CrossRef] [PubMed]
  35. Codolo, G.; Fassan, M.; Munari, F.; Volpe, A.; Bassi, P.; Rugge, M.; Pagano, F.; D’Elios, M.M.; De Bernard, M. HP-NAP Inhibits the Growth of Bladder Cancer in Mice by Activating a Cytotoxic Th1 Response. Cancer Immunol. Immunother. 2012, 61, 31–40. [Google Scholar] [CrossRef] [PubMed]
  36. Davis, L.E.; Shalin, S.C.; Tackett, A.J. Current State of Melanoma Diagnosis and Treatment. Cancer Biol. Ther. 2019, 20, 1366–1379. [Google Scholar] [CrossRef] [PubMed]
  37. Domingues, B.; Lopes, J.M.; Soares, P.; Pópulo, H. Melanoma Treatment in Review. ImmunoTargets Ther. 2018, 7, 35–49. [Google Scholar] [CrossRef] [PubMed]
  38. Henkel, J.S.; Baldwin, M.R.; Barbieri, J.T. Toxins from Bacteria. EXS 2010, 100, 1–29. [Google Scholar] [CrossRef]
  39. Gillet, D.; Barbier, J. Diphtheria Toxin. In The Comprehensive Sourcebook of Bacterial Protein Toxins; Academic Press: Cambridge, MA, USA, 2015; pp. 111–132. [Google Scholar]
  40. Lacy, D.B.; Collier, R.J. Structure and Function of Anthrax Toxin. Curr. Top. Microbiol. Immunol. 2002, 271, 61–85. [Google Scholar] [CrossRef]
  41. Melton-Celsa, A.R. Shiga Toxin (Stx) Classification, Structure, and Function. Microbiol. Spectr. 2014, 2, 2–4. [Google Scholar] [CrossRef]
  42. Vanden Broeck, D.; Horvath, C.; De Wolf, M.J.S. Vibrio Cholerae: Cholera Toxin. Int. J. Biochem. Cell Biol. 2007, 39, 1771–1775. [Google Scholar] [CrossRef]
  43. Johannes, L.; Römer, W. Shiga Toxins—From Cell Biology to Biomedical Applications. Nat. Rev. Microbiol. 2009, 8, 105–116. [Google Scholar] [CrossRef] [PubMed]
  44. Elsland, D.; Neefjes, J. Bacterial Infections and Cancer. EMBO Rep. 2018, 19, e46632. [Google Scholar] [CrossRef] [PubMed]
  45. Matson, V.; Chervin, C.S.; Gajewski, T.F. Cancer and the Microbiome—Influence of the Commensal Microbiota on Cancer, Immune Responses, and Immunotherapy. Gastroenterology 2021, 160, 600–613. [Google Scholar] [CrossRef] [PubMed]
  46. Fettucciari, K.; Macchioni, L.; Davidescu, M.; Scarpelli, P.; Palumbo, C.; Corazzi, L.; Marchegiani, A.; Cerquetella, M.; Spaterna, A.; Marconi, P.; et al. Clostridium Difficile Toxin B Induces Senescence in Enteric Glial Cells: A Potential New Mechanism of Clostridium Difficile Pathogenesis. BBA-Mol. Cell Res. 2018, 1865, 167–4889. [Google Scholar] [CrossRef] [PubMed]
  47. Chandrasekaran, R.; Lacy, D.B. The Role of Toxins in Clostridium Difficile Infection. FEMS Microbiol. Rev. 2017, 41, 723–750. [Google Scholar] [CrossRef] [PubMed]
  48. Khanna, S. Management of Clostridioides Difficile Infection in Patients with Inflammatory Bowel Disease. Intest. Res. 2021, 19, 265–274. [Google Scholar] [CrossRef]
  49. Fettucciari, K.; Ponsini, P.; Gioè, D.; Macchioni, L.; Palumbo, C.; Antonelli, E.; Coaccioli, S.; Villanacci, V.; Corazzi, L.; Marconi, P.; et al. Enteric Glial Cells Are Susceptible to Clostridium Difficile Toxin B. Cell. Mol. Life Sci. 2017, 74, 1527–1551. [Google Scholar] [CrossRef] [PubMed]
  50. Coppé, J.P.; Desprez, P.Y.; Krtolica, A.; Campisi, J. The Senescence-Associated Secretory Phenotype: The Dark Side of Tumor Suppression. Annu. Rev. Pathol. 2010, 5, 99–118. [Google Scholar] [CrossRef]
  51. Schmitt, C.A.; Wang, B.; Demaria, M. Senescence and Cancer—Role and Therapeutic Opportunities. Nat. Rev. Clin. Oncol. 2022, 19, 619–636. [Google Scholar] [CrossRef]
Disclaimer/Publisher’s Note: The statements, opinions and data contained in all publications are solely those of the individual author(s) and contributor(s) and not of MDPI and/or the editor(s). MDPI and/or the editor(s) disclaim responsibility for any injury to people or property resulting from any ideas, methods, instructions or products referred to in the content.

Share and Cite

MDPI and ACS Style

Travaglione, S.; Carlini, F.; Maroccia, Z.; Fabbri, A. Special Issue “Bacterial Toxins and Cancer”. Int. J. Mol. Sci. 2024, 25, 2128. https://doi.org/10.3390/ijms25042128

AMA Style

Travaglione S, Carlini F, Maroccia Z, Fabbri A. Special Issue “Bacterial Toxins and Cancer”. International Journal of Molecular Sciences. 2024; 25(4):2128. https://doi.org/10.3390/ijms25042128

Chicago/Turabian Style

Travaglione, Sara, Francesca Carlini, Zaira Maroccia, and Alessia Fabbri. 2024. "Special Issue “Bacterial Toxins and Cancer”" International Journal of Molecular Sciences 25, no. 4: 2128. https://doi.org/10.3390/ijms25042128

Note that from the first issue of 2016, this journal uses article numbers instead of page numbers. See further details here.

Article Metrics

Back to TopTop