Sign in to use this feature.

Years

Between: -

Subjects

remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline

Journals

Article Types

Countries / Regions

Search Results (169)

Search Parameters:
Keywords = chimeric peptide

Order results
Result details
Results per page
Select all
Export citation of selected articles as:
18 pages, 814 KiB  
Review
Fighting HER2 in Gastric Cancer: Current Approaches and Future Landscapes
by Margherita Ratti, Chiara Citterio, Elena Orlandi, Stefano Vecchia, Elisa Anselmi, Ilaria Toscani, Martina Rotolo, Massimiliano Salati and Michele Ghidini
Int. J. Mol. Sci. 2025, 26(15), 7285; https://doi.org/10.3390/ijms26157285 - 28 Jul 2025
Viewed by 284
Abstract
Gastric cancer (GC) remains a major cause of cancer-related mortality worldwide, with human epidermal growth factor receptor 2 (HER2)-positive disease representing a clinically relevant subset. Trastuzumab combined with chemotherapy is the standard first-line treatment in advanced settings, following the landmark ToGA trial. However, [...] Read more.
Gastric cancer (GC) remains a major cause of cancer-related mortality worldwide, with human epidermal growth factor receptor 2 (HER2)-positive disease representing a clinically relevant subset. Trastuzumab combined with chemotherapy is the standard first-line treatment in advanced settings, following the landmark ToGA trial. However, resistance to trastuzumab has emerged as a significant limitation, prompting the need for more effective second-line therapies. Trastuzumab deruxtecan, a novel antibody–drug conjugate (ADC) composed of trastuzumab linked to a cytotoxic payload, has demonstrated promising efficacy in trastuzumab-refractory, HER2-positive GC, including cases with heterogeneous HER2 expression. Other HER2-targeted ADCs are also under investigation as potential alternatives. In addition, strategies to overcome resistance include HER2-specific immune-based therapies, such as peptide vaccines and chimeric antigen receptor T cell therapies, as well as antibodies targeting distinct HER2 domains or downstream signaling pathways like PI3K/AKT. These emerging approaches aim to improve efficacy in both HER2-high and HER2-low GC. As HER2-targeted treatments evolve, addressing resistance mechanisms and optimizing therapy for broader patient populations is critical. This review discusses current and emerging HER2-directed strategies in GC, focusing on trastuzumab deruxtecan and beyond, and outlines future directions to improve outcomes for patients with HER2-positive GC across all clinical settings. Full article
(This article belongs to the Section Molecular Oncology)
Show Figures

Figure 1

17 pages, 1548 KiB  
Article
CD19-ReTARGTPR: A Novel Fusion Protein for Physiological Engagement of Anti-CMV Cytotoxic T Cells Against CD19-Expressing Malignancies
by Anne Paulien van Wijngaarden, Isabel Britsch, Matthias Peipp, Douwe Freerk Samplonius and Wijnand Helfrich
Cancers 2025, 17(14), 2300; https://doi.org/10.3390/cancers17142300 - 10 Jul 2025
Viewed by 411
Abstract
Background/Objectives: The physiological activation of cytotoxic CD8pos T cells (CTLs) relies on the engagement of the TCR/CD3 complex with cognate peptide-HLA class I (pHLA-I) on target cells, triggering cell lysis with appropriate cytokine release and minimized off-target toxicity. In contrast, current [...] Read more.
Background/Objectives: The physiological activation of cytotoxic CD8pos T cells (CTLs) relies on the engagement of the TCR/CD3 complex with cognate peptide-HLA class I (pHLA-I) on target cells, triggering cell lysis with appropriate cytokine release and minimized off-target toxicity. In contrast, current immunotherapies for CD19-expressing hematological malignancies, such as chimeric antigen receptor (CAR) T cells and bispecific T cell engagers (BiTEs), bypass TCR/pHLA interactions, resulting in CTL hyperactivation and excessive cytokine release, which frequently cause severe immune-related adverse events (irAEs). Thus, there is a pressing need for T cell-based therapies that preserve physiological activation while maintaining antitumor efficacy. Methods: To address this, we developed CD19-ReTARGTPR, a novel fusion protein consisting of the immunodominant cytomegalovirus (CMV) pp65-derived peptide TPRVTGGAM (TPR) covalently presented by a soluble HLA-B*07:02/β2-microglobulin complex fused to a high-affinity CD19-targeting Fab antibody fragment. The treatment of CD19-expressing cancer cells with CD19-ReTARGTPR makes them recognizable for pre-existing anti-CMVpp65 CTLs via physiological TCR-pHLA engagement. Results: Our preclinical data demonstrate that CD19-ReTARGTPR efficiently redirects anti-CMV CTLs to eliminate CD19-expressing cancer cells, including both established cell lines and primary chronic lymphocytic leukemia (CLL) cells. Unlike CD19-directed CAR T cells or the CD19/CD3 BiTE blinatumomab, CD19-ReTARGTPR mediated robust cytotoxic activity without triggering supraphysiological cytokine release. Importantly, this approach retained efficacy even against cancer cells with low CD19 expression. Conclusions: In summary, we provide a robust proof-of-concept study and show that CD19-ReTARGTPR offers a promising alternative strategy for T cell redirection, enabling the selective and effective killing of CD19-expressing malignancies while minimizing cytokine-driven toxicities through physiological CTL activation pathways. Full article
(This article belongs to the Special Issue New Insights of Hematology in Cancer)
Show Figures

Graphical abstract

14 pages, 1004 KiB  
Article
Designing New Chimeric Proline-Rich Antimicrobial Peptides to Enhance Efficacy Toward the ESKAPE+E: Beyond Sequence Extension
by Adriana Di Stasi, Luigi de Pascale, Martino Morici, Daniel N. Wilson, Marco Scocchi and Mario Mardirossian
Biomolecules 2025, 15(6), 776; https://doi.org/10.3390/biom15060776 - 27 May 2025
Viewed by 621
Abstract
Proline-rich antimicrobial peptides (PrAMPs) primarily exert their antimicrobial effects intracellularly, inhibiting protein synthesis. B7-005, a synthetic 16-amino acid PrAMP, has a broader antimicrobial spectrum compared to native counterparts, despite shorter PrAMPs typically exhibiting reduced activity. This study aimed to enhance B7-005’s potency by [...] Read more.
Proline-rich antimicrobial peptides (PrAMPs) primarily exert their antimicrobial effects intracellularly, inhibiting protein synthesis. B7-005, a synthetic 16-amino acid PrAMP, has a broader antimicrobial spectrum compared to native counterparts, despite shorter PrAMPs typically exhibiting reduced activity. This study aimed to enhance B7-005’s potency by extending it with 6 or 11 amino acids derived from the C-terminal sequences of cetacean Tur1A and Lip1 PrAMPs, as well as bovine Bac7(1-35). Six chimeric derivatives were evaluated for antimicrobial and bactericidal potency, cytotoxicity, bacterial membrane permeabilization, and in vitro inhibition of protein synthesis. Extending B7-005 with sequences from other PrAMPs increased its activity against most ESKAPE+E pathogens, reducing minimum inhibitory concentration (MIC) values by 2- to 8-fold, with notable differences among bacterial species, without increasing cytotoxicity toward the A549 cell line. All chimeras retained the ability to inhibit protein synthesis in Escherichia coli and to modestly perturb the E. coli membranes like B7-005. These novel chimeric PrAMPs, particularly the 22-mer derivatives, hold promise for developing new antimicrobial agents. The study also highlights variability in bacterial responses to PrAMPs and underscores how minor sequence differences can significantly impact efficacy against specific microorganisms. PrAMPs thus represent a valuable scaffold to rationally design derivatives targeting high-priority pathogens. Full article
(This article belongs to the Special Issue State of the Art and Perspectives in Antimicrobial Peptides)
Show Figures

Graphical abstract

24 pages, 754 KiB  
Review
Chimeric Autoantibody Receptor- and/or Peptide-MHC-Based CAR Therapies for Targeted Elimination of Antigen-Specific B or T Cells in Hypersensitivity Disorders Such as Allergies and Autoimmune Diseases
by Isidora Protić-Rosić, Al Nasar Ahmed Sehgal, Sebastian Wrighton, Birgit Heller and Winfried F. Pickl
Cells 2025, 14(10), 753; https://doi.org/10.3390/cells14100753 - 21 May 2025
Viewed by 987
Abstract
Hypersensitivity reactions are dysregulated and potentially devastating immune responses, characterized by a tendency to become chronic. They target either self-proteins or harmless foreign proteins and are driven by both T and B cells. Although numerous symptomatic treatment options for hypersensitivity reactions have been [...] Read more.
Hypersensitivity reactions are dysregulated and potentially devastating immune responses, characterized by a tendency to become chronic. They target either self-proteins or harmless foreign proteins and are driven by both T and B cells. Although numerous symptomatic treatment options for hypersensitivity reactions have been established over recent decades, only a few antigen-specific, causal approaches capable of specifically targeting the pathogenic autoreactive T and/or B cells have been developed. Among these are cell-based treatment modalities involving chimeric antigen receptor (CAR)- or chimeric autoantibody-receptor (CAAR)-expressing cells. These therapies utilize B- or T-cell antigens, presented as B-cell epitopes or peptide-major histocompatibility complexes (pMHCs) to serve as bait. The latter are coupled to potent activation domains derived from the TCR/CD3 complex itself, such as the zeta or CD3 chains, as well as domains from bona fide co-stimulatory molecules (e.g., CD28, 4-1BB). Recent in vitro and in vivo studies have demonstrated the therapeutic potential of these ATMP-based strategies in eliminating autoreactive lymphocytes and alleviating hypersensitivity reactions. This systematic review provides a comprehensive overview of the current status of antigen-specific CAR and CAAR T-cell therapies, highlighting novel directions as well as the ongoing challenges within this promising research field. Full article
Show Figures

Figure 1

21 pages, 312 KiB  
Review
Update: Immunotherapeutic Strategies in HPV-Associated Head and Neck Squamous Cell Carcinoma
by Fangdi Sun and A. Dimitrios Colevas
Viruses 2025, 17(5), 712; https://doi.org/10.3390/v17050712 - 16 May 2025
Viewed by 961
Abstract
The incidence of human papillomavirus (HPV)-associated oropharyngeal squamous cell carcinoma (OPSCC) has increased substantially over the past three decades, and since 2017, it has been recognized in the AJCC staging system as distinct from its HPV-negative counterpart. The underlying mechanisms of HPV-associated carcinogenesis, [...] Read more.
The incidence of human papillomavirus (HPV)-associated oropharyngeal squamous cell carcinoma (OPSCC) has increased substantially over the past three decades, and since 2017, it has been recognized in the AJCC staging system as distinct from its HPV-negative counterpart. The underlying mechanisms of HPV-associated carcinogenesis, tumor microenvironment, and host immune response represent opportunities for therapeutic development. While anti-PD-1 immunotherapy is now part of standard treatment for recurrent or metastatic head and neck squamous cell carcinoma (HNSCC) in general, there are no established immunotherapeutic strategies specifically for HPV-related HNSCC. In this context, multiple emerging approaches are being actively studied—among these are therapeutic vaccines with or without anti-PD-(L)1 adjuvants, peptide–HLA-based immunotherapeutic platforms, and adoptive cell therapies including tumor-infiltrating lymphocytes (TILs), T-cell receptor (TCR) therapy, and chimeric antigen receptor (CAR) T-cell therapy. Beyond further maturation of these novel immunotherapeutic strategies, additional work is needed to delineate the optimal disease state of application (localized versus recurrent/metastatic), as well as in the development of small molecule inhibitors targeting HPV-specific mechanisms of viral oncogenesis. Full article
(This article belongs to the Special Issue Advancements in Immunotherapy for Human Papillomavirus)
17 pages, 1223 KiB  
Article
Dynamics of IgM and IgA Antibody Response Profile Against Vibrio cholerae Toxins A, B, and P
by Salvatore Giovanni De-Simone, Paloma Napoleão-Pêgo, Guilherme Curty Lechuga, Joao Pedro Rangel Silva Carvalho, Sergian Vianna Cardozo, Alexandre Oliveira Saisse, Carlos Medicis Morel, David William Provance and Flavio Rocha da Silva
Int. J. Mol. Sci. 2025, 26(8), 3507; https://doi.org/10.3390/ijms26083507 - 9 Apr 2025
Cited by 1 | Viewed by 583
Abstract
The first immune response controls many bacterial and viral inflammatory diseases. Oral immunization with cholera toxin (CT) elicits antibodies and can prevent cholerae in endemic environments. While the IgG immune response to the toxin is well-documented, the IgA and IgM epitopes responsible for [...] Read more.
The first immune response controls many bacterial and viral inflammatory diseases. Oral immunization with cholera toxin (CT) elicits antibodies and can prevent cholerae in endemic environments. While the IgG immune response to the toxin is well-documented, the IgA and IgM epitopes responsible for the initial immune reaction to the toxin remained uncharted. In this study, our objective was to identify and characterize immunologically and structurally these IgA and IgM epitopes. We conducted SPOT synthesis to create two libraries, each containing one hundred twenty-two 15-mer peptides, encompassing the entire sequence of the three chains of the CT protein. We could map continuous IgA and IgM epitopes by testing these membrane-bound peptides with sera from mice immunized with an oral vaccine (Schankol™). Our approach involved topological studies, peptide synthesis, and the development of an ELISA. We successfully identified seven IgA epitopes, two in CTA, two in CTB, and three in protein P. Additionally, we discovered eleven IgM epitopes, all situated within CTA. Three IgA-specific and three IgM-specific epitopes were synthesized as MAP4 and validated using ELISA. We then used two chimeric 45-mer peptides, which included these six epitopes, to coat ELISA plates and screened them with sera from immunized mice. This yielded sensitivities and specificities of 100%. Our findings have unveiled a significant collection of IgA and IgM-specific peptide epitopes from cholera toxins A, B, and P. These epitopes, along with those IgG previously identified by our group, reflect the immunoreactivity associated with the dynamic of the immunoglobulins switching associated with the cholera toxin vaccination. Full article
(This article belongs to the Section Molecular Biophysics)
Show Figures

Figure 1

30 pages, 2105 KiB  
Article
The Chimeric Peptide (GEP44) Reduces Body Weight and Both Energy Intake and Energy Expenditure in Diet-Induced Obese Rats
by Matvey Goldberg, James E. Blevins, Tami Wolden-Hanson, Clinton T. Elfers, Kylie S. Chichura, Emily F. Ashlaw, Laura J. den Hartigh, Christian L. Roth and Robert P. Doyle
Int. J. Mol. Sci. 2025, 26(7), 3032; https://doi.org/10.3390/ijms26073032 - 26 Mar 2025
Viewed by 756
Abstract
We recently reported that a chimeric peptide (GEP44) targeting the glucagon-like peptide-1 receptor (GLP-1R) and neuropeptide Y1- and Y2- receptors decreased body weight (BW), energy intake, and core temperature in diet-induced obese (DIO) male and female mice. In the current study, we tested [...] Read more.
We recently reported that a chimeric peptide (GEP44) targeting the glucagon-like peptide-1 receptor (GLP-1R) and neuropeptide Y1- and Y2- receptors decreased body weight (BW), energy intake, and core temperature in diet-induced obese (DIO) male and female mice. In the current study, we tested the hypothesis that the strong reduction in body weight in response to GEP44 is partially related to the stimulation of energy expenditure (EE). To test this, rats were maintained on a high fat diet (HFD) for at least 4 months to elicit DIO prior to undergoing a sequential 2-day vehicle period, 2-day GEP44 (50 nmol/kg) period, and a minimum 2-day washout period, and detailed measures of energy homeostasis. GEP44 (50 nmol/kg) reduced EE (indirect calorimetry), respiratory exchange ratio (RER), core temperature, activity, energy intake, and BW in male and female rats. As in our previous study in mice, GEP44 reduced BW in male and female HFD-fed rats by 3.8 ± 0.2% and 2.3 ± 0.4%, respectively. These effects appear to be mediated by increased lipid oxidation and reductions in energy intake as GEP44 reduced RER and cumulative energy intake in male and female HFD-fed rats. The strong reduction in body weight in response to GEP44 is related to a robust reduction in energy intake, but not to the stimulation of EE. The paradoxical finding that GEP44 reduced EE might be secondary to a reduction in diet-induced thermogenesis or might indicate an important mechanism to limit the overall efficacy of GEP44 to prevent further weight loss. Full article
(This article belongs to the Special Issue The Effect of Food-Derived Compounds on Brown Fat Cell Function)
Show Figures

Figure 1

21 pages, 2530 KiB  
Review
Pathogenesis and Immunomodulation of Urinary Tract Infections Caused by Uropathogenic Escherichia coli
by J. David García-García, Laura M. Contreras-Alvarado, Ariadnna Cruz-Córdova, Rigoberto Hernández-Castro, Marcos Flores-Encarnacion, Sandra Rivera-Gutiérrez, José Arellano-Galindo, Sara A. Ochoa and Juan Xicohtencatl-Cortes
Microorganisms 2025, 13(4), 745; https://doi.org/10.3390/microorganisms13040745 - 26 Mar 2025
Viewed by 2330
Abstract
Urinary tract infections (UTIs) are a leading cause of illness in children and adults of all ages, with uropathogenic Escherichia coli (UPEC) being the primary agent responsible. During colonization and subsequent infection of the urinary tract (UT), UPEC requires the expression of genes [...] Read more.
Urinary tract infections (UTIs) are a leading cause of illness in children and adults of all ages, with uropathogenic Escherichia coli (UPEC) being the primary agent responsible. During colonization and subsequent infection of the urinary tract (UT), UPEC requires the expression of genes associated with virulence, such as those that encode the fimbrial adhesins FimH, PapG, and CsgA, as well as the presence of the TosA protein and the flagellar appendages of the bacteria. However, for colonization and infection to be successful, UPEC must overcome the host’s immunological barriers, such as physical barriers, expressed peptides and proteins, and immune cells found in the UT. In this context, the UT functions as an integral system where these factors act to prevent the colonization of uropathogens. Significant genetic diversity exists among UPEC strains, and the clonal complex ST131 represents one of the key lineages. This lineage has a high content of virulence genes, multiple mechanisms of antibiotic resistance, and a high frequency of extended-spectrum β-lactamases (ESBLs). New knowledge regarding protein structures known as adhesins and their role in the infection process can help identify therapeutic targets and aid in the design of vaccines. These vaccines could be based on the development of chimeric fusion proteins (FimH + CsgA + PapG), which may significantly reduce the incidence of UTIs in pediatric and adult patients. Full article
(This article belongs to the Special Issue Pathogenic Mechanisms of Bacterial Infections)
Show Figures

Figure 1

11 pages, 1574 KiB  
Article
CLEC3A-Derived Antimicrobial Peptides Reduce Staphylococcus aureus Bacterial Counts in an In Vivo Biomaterial-Associated Infection Mouse Model
by Denise Meinberger, Gabriele Hermes, Bent Brachvogel, Gerhard Sengle, Dzemal Elezagic, Annika Roth, Johannes Ruthard, Thomas Streichert and Andreas R. Klatt
Pharmaceutics 2025, 17(2), 234; https://doi.org/10.3390/pharmaceutics17020234 - 12 Feb 2025
Viewed by 901
Abstract
Background/Objectives: Biomaterials are an essential part of healthcare for both diagnostic and therapeutic procedures. Although some biomaterials possess antimicrobial properties, introducing biomaterial into the body may lead to infections due to bacterial adhesion on their surfaces and still poses a major clinical problem. [...] Read more.
Background/Objectives: Biomaterials are an essential part of healthcare for both diagnostic and therapeutic procedures. Although some biomaterials possess antimicrobial properties, introducing biomaterial into the body may lead to infections due to bacterial adhesion on their surfaces and still poses a major clinical problem. Peptides derived from the human cartilage-specific C-type lectin domain family 3 member A (CLEC3A) show a potent antimicrobial effect. In addition, coating titanium, a commonly used prosthetic material, with the CLEC3A-derived AMPs HT-47 and WRK-30 greatly reduces the number of adherent bacteria in vitro. The aim of this study was to evaluate the effectiveness of CLEC3A-derived peptides HT-47 and WRK-30 in reducing bacterial adhesion and mitigating infection in vivo in a murine biomaterial-associated infection model. Methods: To do so, an in vivo mouse infection model was used, where titanium plates—either uncoated or coated with chimeric CLEC3A-derived peptides TiBP-HT-47 and TiBP-WRK-30—were implanted subcutaneously into mice. This was followed by the introduction of Staphylococcus aureus bacterial cultures to induce a biomaterial-associated infection. After 24 h, the titanium plates, surrounding tissue, and mice blood samples were investigated. Results: CLEC3A-coated titanium plates lead to a significantly lower bacterial count than uncoated ones. Additionally, they prevent the infection from spreading to the surrounding tissue. Moreover, mice with CLEC3A-coated implants display lower IL-6 serum levels and therefore decreased systemic inflammation. Conclusions: In conclusion, in this biomaterial-associated infection mouse-model, CLEC3A-derived peptides show in vivo antimicrobial activity by reducing bacterial burden on biomaterial and wound tissue and decreasing systemic inflammation, making them promising candidates for clinical applications. Full article
Show Figures

Graphical abstract

15 pages, 20405 KiB  
Article
Relative Quantitation of EFNA1 Expression in Mouse Heart Tissue Histologic Sections Using MALDI-MSI
by Maria Torres, Laura Gruer, Smrithi Valsaraj, Shaun Reece, Jeremy Prokop, Tonya Zeczycki, Cameron Taylor, Taylor Byers, William Cruz, Kim Kew, Lisandra de Castro Braz and Jitka Virag
Int. J. Mol. Sci. 2025, 26(4), 1398; https://doi.org/10.3390/ijms26041398 - 7 Feb 2025
Viewed by 968
Abstract
EFNA1 (ephrinA1), a highly expressed tyrosine kinase receptor-ligand in healthy cardiomyocytes, is reduced following myocardial infarction (MI). A single intramyocardial injection of chimeric EFNA1-Fc at the time of ischemia mitigates the injury in both reperfused and non-reperfused mouse myocardium by reducing apoptosis, necrosis, [...] Read more.
EFNA1 (ephrinA1), a highly expressed tyrosine kinase receptor-ligand in healthy cardiomyocytes, is reduced following myocardial infarction (MI). A single intramyocardial injection of chimeric EFNA1-Fc at the time of ischemia mitigates the injury in both reperfused and non-reperfused mouse myocardium by reducing apoptosis, necrosis, and inflammation. Recently, we have successfully imaged and qualitatively identified endogenous EFNA1 pre- and post-MI using matrix-assisted laser desorption ionization mass spectrometry imaging (MALDI-MSI) coupled with a time-of-flight mass spectrometer (MALDI/TOF MS). Building on our previous work, we are currently focused on understanding and characterizing EFNA1’s role in cardiac tissue by developing an integrated quantitative method to determine endogenous levels of EFNA1 using MALDI-MSI technologies. Herein, we have optimized a method for the relative quantitation of endogenous tryptic EFNA1 peptides detected in the murine heart as compared with routine western blotting. In healthy myocardium, there was approximately 50 ng of endogenous EFNA1 per section of 9.43 mm3 tissue, or roughly 12 pg/µg of homogenized tissue. MALDI-MSI thus provides a tool for determining the anatomical distribution and relative quantitation of endogenous EFNA1 in cardiac tissue. Future applications of these tools will allow us to investigate the dynamic changes in EFNA1 expression profile that occur in pathological states such as myocardial infarction and upon therapeutic treatments. Full article
(This article belongs to the Special Issue Research Progress on the Mechanism and Treatment of Cardiomyopathy)
Show Figures

Figure 1

19 pages, 3649 KiB  
Article
Evaluating TcAs for Use in Biotechnology Applications
by Cole L. Martin, John H. Hill, Brian D. Wright, Solana R. Fernandez, Aubrey L. Miller, Karina J. Yoon, Suzanne E. Lapi and Stephen G. Aller
BioTech 2025, 14(1), 5; https://doi.org/10.3390/biotech14010005 - 25 Jan 2025
Viewed by 1502
Abstract
ABC toxin complexes (Tcs) are tripartite complexes that come together to form nano-syringe-like translocation systems. ABC Tcs are often compared with Bacillus thuringiensis (Bt) toxins, and as such, they have been highly studied as a potential novel pesticide to combat growing insect resistance. [...] Read more.
ABC toxin complexes (Tcs) are tripartite complexes that come together to form nano-syringe-like translocation systems. ABC Tcs are often compared with Bacillus thuringiensis (Bt) toxins, and as such, they have been highly studied as a potential novel pesticide to combat growing insect resistance. Moreover, it is possible to substitute the cytotoxic hypervariable region with alternative peptides, which promise potential use as a novel peptide delivery system. These toxins possess the unique ability to form active chimeric holotoxins across species and display the capability to translocate a variety of payloads across membrane bilayers. Additionally, mutagenesis on the linker region and the receptor binding domains (RBDs) show that mutations do not inherently cause a loss of functionality for translocation. For these reasons, Tcs have emerged as an ideal candidate for targeted protein engineering. However, elucidation of the specific function of each RBD in relation to target receptor recognition currently limits the use of a rational design approach with any ABC Tc. Additionally, there is a distinct lack of targeting and biodistribution data for many Tcs among mammals and mammalian cell lines. Here, we outline two separate strategies for modifying the targeting capabilities of the A subunit (TcA) from Xenorhabdus nematophilus, Xn-XptA2. We identify novel structural differences that make Xn-XptA2 different than other characterized TcAs and display the modular capabilities of substituting RBDs from alternative TcAs into the Xn-XptA2 scaffold. Finally, we show the first, to our knowledge, biodistribution data of any TcA in mice. Full article
Show Figures

Figure 1

16 pages, 3927 KiB  
Article
Activity of Potassium Channel BmK-NSPK Inhibitor Regulated by Basic Amino Acid Residues: Novel Insight into the Diverse Peptide Pharmacology
by Zheng Zuo, Xuhua Yang, Haozhen Zhang, Chenhu Qin, Zhijian Cao and Yingliang Wu
Molecules 2025, 30(3), 450; https://doi.org/10.3390/molecules30030450 - 21 Jan 2025
Viewed by 763
Abstract
The molecular interactions between venomous peptides and potassium channels have extensively enriched the knowledge of diverse peptide pharmacology, and the in-depth understanding of general features of the various peptide functions remains a formidable challenge. In this work, the role of peptide basic residues [...] Read more.
The molecular interactions between venomous peptides and potassium channels have extensively enriched the knowledge of diverse peptide pharmacology, and the in-depth understanding of general features of the various peptide functions remains a formidable challenge. In this work, the role of peptide basic residues in peptide pharmacology was first investigated. Although the venomous BmK-NSPK peptide had the critically conserved functional residues occurring in its similar and potent potassium channel-inhibiting peptides, it was a remarkably weak inhibitor of potassium channels due to fewer basic residues. Additionally, 1 μM BmK-NSPK only inhibited 1.2 ± 1.0%, 1.7 ± 0.70%, 2.3 ± 0.49% and 5.4 ± 0.70% of hKv1.1, hKv1.2, hKv1.3 and hKv1.6 channel currents. The introduction of one or two basic residues in BmK-NSPK-I15K, BmK-NSPK-I18K, BmK-NSPK-I26K and BmK-NSPK-I18K/I26K could not improve BmK-NSPK activity. The modifications of more than three basic residues were found to continuously improve BmK-NSPK activity, and the corresponding BmK-NSPK-7K and BmK-NSPK-8K mutants could effectively inhibit hKv1.3 channel with IC50 values of 2.04 ± 0.68 nM and 21.5 ± 1.99 nM, respectively. Also, 1 μM BmK-NSPK-7K and BmK-NSPK-8K mutants could inhibit 84.1 ± 7.0% and 84.3 ± 1.8% of hKv1.1 channel currents. In addition, BmK-NSPK-7K and BmK-NSPK-8K mutants were found to differentially inhibit hKv1.6 and chimeric hKv1.3 channels. These findings first highlight the critical role of basic residues in the activity of potassium channel peptide inhibitors and provide novel insight into the diverse peptide pharmacology. Full article
Show Figures

Figure 1

12 pages, 2281 KiB  
Article
CD40 Ligand Potentiates Immunogenecity of Mycoplasma pneumoniae Subunit Vaccine Candidate in a Murine Model
by Jinqi Shu, Gaojian Li, Jianhong Shu, Huapeng Feng and Yulong He
Curr. Issues Mol. Biol. 2025, 47(1), 37; https://doi.org/10.3390/cimb47010037 - 9 Jan 2025
Viewed by 1083
Abstract
Mycoplasma hyopneumoniae (Mhp) infection severely affects the daily weight gain and feed-to-meat ratio of pigs, while secondary infections with other pathogens can further lead to increased mortality, causing significant economic losses to the pig industry. CD40L is a molecular adjuvant that enhances the [...] Read more.
Mycoplasma hyopneumoniae (Mhp) infection severely affects the daily weight gain and feed-to-meat ratio of pigs, while secondary infections with other pathogens can further lead to increased mortality, causing significant economic losses to the pig industry. CD40L is a molecular adjuvant that enhances the cellular and humoral immune responses to vaccines. In this study, the CD40L peptide was fused to the C-terminus of the chimeric P97R1P46P42 protein by genetic engineering using the pFastBac Dual vector. The recombinant chimeric protein P97R1P46P42 and its fusion P97R1P46P42-CD40L were expressed in Sf9 cells and purified. Mice were immunized with P97R1P46P42 or its fusion protein. Seppic ISA 201 emulsified protein, conventional Mhp vaccine and PBS control groups were included. Immunogenecity was assessed by specific IgG antibody response, splenic lymphocyte proliferation, and cytokine IL-4 and IFN-γ levels. We found that CD40L fusion significantly enhanced specific antibody response, lymphocyte proliferation and IL-4 level in the immunized mouse sera as compared to the P97R1P46P42 or conventional vaccine group. This study provides clear evidence that CD40L potentiates the humoral and cellular immune responses to the Mhp chimeric protein P97R1P46P42 in the mouse model. This CD40L-fused chimeric protein could be a MPS subunit vaccine candidate to be tested for its efficacy in pigs in response to challenges with pathogenic Mycoplasma hyopneumoniae strain(s). Full article
Show Figures

Figure 1

26 pages, 1514 KiB  
Review
Challenges and Insights in Absolute Quantification of Recombinant Therapeutic Antibodies by Mass Spectrometry: An Introductory Review
by Sarah Döring, Michael G. Weller, Yvonne Reinders, Zoltán Konthur and Carsten Jaeger
Antibodies 2025, 14(1), 3; https://doi.org/10.3390/antib14010003 - 7 Jan 2025
Cited by 1 | Viewed by 2982
Abstract
This review describes mass spectrometry (MS)-based approaches for the absolute quantification of therapeutic monoclonal antibodies (mAbs), focusing on technical challenges in sample treatment and calibration. Therapeutic mAbs are crucial for treating cancer and inflammatory, infectious, and autoimmune diseases. We trace their development from [...] Read more.
This review describes mass spectrometry (MS)-based approaches for the absolute quantification of therapeutic monoclonal antibodies (mAbs), focusing on technical challenges in sample treatment and calibration. Therapeutic mAbs are crucial for treating cancer and inflammatory, infectious, and autoimmune diseases. We trace their development from hybridoma technology and the first murine mAbs in 1975 to today’s chimeric and fully human mAbs. With increasing commercial relevance, the absolute quantification of mAbs, traceable to an international standard system of units (SI units), has attracted attention from science, industry, and national metrology institutes (NMIs). Quantification of proteotypic peptides after enzymatic digestion using high-performance liquid chromatography-tandem mass spectrometry (HPLC-MS/MS) has emerged as the most viable strategy, though methods targeting intact mAbs are still being explored. We review peptide-based quantification, focusing on critical experimental steps like denaturation, reduction, alkylation, choice of digestion enzyme, and selection of signature peptides. Challenges in amino acid analysis (AAA) for quantifying pure mAbs and peptide calibrators, along with software tools for targeted MS data analysis, are also discussed. Short explanations within each chapter provide newcomers with an overview of the field’s challenges. We conclude that, despite recent progress, further efforts are needed to overcome the many technical hurdles along the quantification workflow and discuss the prospects of developing standardized protocols and certified reference materials (CRMs) for this goal. We also suggest future applications of newer technologies for absolute mAb quantification. Full article
Show Figures

Figure 1

17 pages, 2636 KiB  
Article
Development of Peptide Mimics of the Human Acetylcholine Receptor Main Immunogenic Region for Treating Myasthenia Gravis
by Vu B. Trinh and Robert H. Fairclough
Int. J. Mol. Sci. 2025, 26(1), 229; https://doi.org/10.3390/ijms26010229 - 30 Dec 2024
Cited by 1 | Viewed by 981
Abstract
We have designed and produced 39 amino acid peptide mimics of the Torpedo and human acetylcholine receptors’ (AChRs) main immunogenic regions (MIRs). These conformationally sensitive regions consist of three non-contiguous segments of the AChR α-subunits and are the target of 50–70% of the [...] Read more.
We have designed and produced 39 amino acid peptide mimics of the Torpedo and human acetylcholine receptors’ (AChRs) main immunogenic regions (MIRs). These conformationally sensitive regions consist of three non-contiguous segments of the AChR α-subunits and are the target of 50–70% of the anti-AChR autoantibodies (Abs) in human myasthenic serum and in the serum of rats with a model of that disease, experimental autoimmune myasthenia gravis (EAMG), induced by immunizing the rats with the Torpedo electric organ AChR. These MIR segments covalently joined together bind a significant fraction of the monoclonal antibodies (mAbs) raised in rats against electric organ AChR. Many of these mAbs cross react with the rat neuromuscular AChR MIR and induce myasthenic symptoms when injected into naïve rats. The human MIR mimic peptide (H39MIR) is evolutionarily related to that of the Torpedo electric organ MIR mimic peptide (T39MIR) with eight amino acid differences between the two MIR mimics. The mAbs raised to the electric organ AChR MIR cross react with the human and scores of other species’ neuromuscular AChRs. However, the mAbs do not cross react with the H39MIR mimic attached to the N-terminus of an intein–chitin-binding domain (H39MIR-IChBD) even though they do bind to the T39MIR-IChBD construct. To account for this difference in binding anti-MIR mAbs, each of the eight human amino acids was substituted individually into the T39MIR-IChBD, and four of them were found to weaken mAb recognition. Substituting the corresponding four Torpedo amino acids individually and in combination into the homologous positions in H39MIR-IChBD makes chimeric human MIR mimic peptides (T/H39MIR), some of which bind anti-MIR mAbs and anti-MIR Abs from rat EAMG and human MG sera. The best mAb binding chimeric peptide constructs may potentially serve as the basis of a diagnostic anti-MIR Ab titer assay that is both prognostic and predictive of disease severity. Furthermore, the best peptides may also serve as the targeting element of a non-steroidal antigen-specific treatment of MG to remove anti-AChR MIR Abs, either as fused to the N-terminals of the human immunoglobin Fc fragment or as the targeting component of a T cell chimeric autoantibody receptor (CAAR) directed to anti-MIR memory B cells for elimination. Full article
(This article belongs to the Special Issue Autoimmune Diseases: From Molecular Basis to Therapy)
Show Figures

Figure 1

Back to TopTop