1
|
Gaonkar RH, Bailly T, Millul J, Mansi R, Harms M, Münch J, Fani M. Improving Affinity while Reducing Kidney Uptake of CXCR4-Targeting Radioligands Derived from the Endogenous Antagonist EPI-X4. ChemMedChem 2025; 20:e202400773. [PMID: 39782735 DOI: 10.1002/cmdc.202400773] [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: 09/28/2024] [Revised: 12/20/2024] [Accepted: 01/08/2025] [Indexed: 01/12/2025]
Abstract
The C-X-C chemokine receptor 4 (CXCR4) is highly upregulated in most cancers, making it an ideal target for delivering radiation therapy to tumors. We previously demonstrated the feasibility of targeting CXCR4 in vivo using a radiolabeled derivative of EPI-X4, an endogenous CXCR4 antagonist, named DOTA-K-JM#173. However, this derivative showed undesirable accumulation in the kidneys, which would limit its clinical use. In this study, we identified that removing a positive charge from the peptide sequence significantly reduced renal uptake. We evaluated a series of optimized derivatives lacking this positive charge, in vitro and in vivo in a xenografted athymic nude mice model, after radiolabeling with 177Lu. The most promising derivatives were further assessed in vivo after 68Ga labeling. Among them, we identified DOTA-JM#173 and D-L1-DOTA-JM#173, where the D-Ile1 was replaced by D-Leu1, two optimized derivatives with a lysine residue removed. These two molecules represent the most advanced DOTA-conjugated ligands derived from EPI-X4 for CXCR4-directed theranostic applications, offering enhanced potential for targeted cancer treatment.
Collapse
Affiliation(s)
- Raghuvir H Gaonkar
- Division of Radiopharmaceutical Chemistry, Department Theragnostics, University Hospital Basel, Petersgraben 4, 4031 Basel, Switzerland
| | - Thibaud Bailly
- Division of Radiopharmaceutical Chemistry, Department Theragnostics, University Hospital Basel, Petersgraben 4, 4031 Basel, Switzerland
| | - Jacopo Millul
- Division of Radiopharmaceutical Chemistry, Department Theragnostics, University Hospital Basel, Petersgraben 4, 4031 Basel, Switzerland
| | - Rosalba Mansi
- Division of Radiopharmaceutical Chemistry, Department Theragnostics, University Hospital Basel, Petersgraben 4, 4031 Basel, Switzerland
| | - Mirja Harms
- Institute of Molecular Virology, Ulm University Medical Center Meyerhofstraße 1,89081, Ulm, Germany
| | - Jan Münch
- Institute of Molecular Virology, Ulm University Medical Center Meyerhofstraße 1,89081, Ulm, Germany
| | - Melpomeni Fani
- Division of Radiopharmaceutical Chemistry, Department Theragnostics, University Hospital Basel, Petersgraben 4, 4031 Basel, Switzerland
| |
Collapse
|
2
|
Khunti N, Kumar M, Datta M, Harelimana JDD, Harms M, Albers D, Kirchhoff F, Münch J, Stenger S, Buske C, Maity PC. CXCR4 Inhibition Enhances the Efficacy of CD19 Monoclonal Antibody-Mediated Extermination of B-Cell Lymphoma. Int J Mol Sci 2025; 26:2024. [PMID: 40076664 PMCID: PMC11899823 DOI: 10.3390/ijms26052024] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2024] [Revised: 02/17/2025] [Accepted: 02/21/2025] [Indexed: 03/14/2025] Open
Abstract
CD19 and CXCR4 are pivotal regulators of B-cell activation and migration, respectively. Specifically, CXCR4 signaling critically influences the dissemination of various malignant B cells through constitutive activation and aberrant expression. This study explores the interaction between CD19 and CXCR4 signaling in the context of B-cell lymphomas, particularly focusing on diffuse large B-cell lymphoma (DLBCL) and Waldenström Macroglobulinemia (WM). We assessed the roles of CD19 in survival and CXCL12-induced migration by using knockout (KO) cells of DLBCL and WM origin alongside evaluating the impact of CD19 monoclonal antibodies (mAbs) on antibody-dependent cell-mediated cytotoxicity (ADCC). Our results highlight that CD19 is important for survival and CXCL12-induced migration, and mAbs variably increase CXCL12-induced migration and enhance ADCC. Additionally, we demonstrate that the endogenous peptide inhibitor of the CXCR4 (EPI-X4) derivative JM#21 effectively inhibits CD19-mediated migration enhancement and promotes ADCC, thereby augmenting the therapeutic efficacy of CD19 mAb-based immunotherapy in lymphoma models. Our study underscores the potential of targeting both CD19 and CXCR4 to refine therapeutic strategies for treating B-cell malignancies, suggesting a synergistic approach could improve clinical outcomes in WM treatment.
Collapse
MESH Headings
- Receptors, CXCR4/antagonists & inhibitors
- Receptors, CXCR4/metabolism
- Antigens, CD19/immunology
- Antigens, CD19/metabolism
- Humans
- Antibodies, Monoclonal/pharmacology
- Cell Line, Tumor
- Cell Movement/drug effects
- Lymphoma, Large B-Cell, Diffuse/immunology
- Lymphoma, Large B-Cell, Diffuse/drug therapy
- Lymphoma, Large B-Cell, Diffuse/metabolism
- Lymphoma, Large B-Cell, Diffuse/pathology
- Lymphoma, B-Cell/drug therapy
- Lymphoma, B-Cell/metabolism
- Lymphoma, B-Cell/immunology
- Lymphoma, B-Cell/pathology
- Antibody-Dependent Cell Cytotoxicity/drug effects
- Chemokine CXCL12/metabolism
- Animals
- Waldenstrom Macroglobulinemia/drug therapy
- Waldenstrom Macroglobulinemia/immunology
- Waldenstrom Macroglobulinemia/metabolism
- Waldenstrom Macroglobulinemia/pathology
- Mice
- Signal Transduction/drug effects
- Antineoplastic Agents, Immunological/pharmacology
- Antineoplastic Agents, Immunological/therapeutic use
Collapse
Affiliation(s)
- Nupur Khunti
- Institute of Experimental Cancer Research, Ulm University Medical Center, 89081 Ulm, Germany (M.K.)
| | - Manish Kumar
- Institute of Experimental Cancer Research, Ulm University Medical Center, 89081 Ulm, Germany (M.K.)
| | - Moumita Datta
- Institute of Immunology, Ulm University Medical Center, 89081 Ulm, Germany;
| | | | - Mirja Harms
- Institute of Molecular Virology, Ulm University Medical Center, 89081 Ulm, Germany
| | - Dan Albers
- Institute of Molecular Virology, Ulm University Medical Center, 89081 Ulm, Germany
| | - Frank Kirchhoff
- Institute of Molecular Virology, Ulm University Medical Center, 89081 Ulm, Germany
| | - Jan Münch
- Institute of Molecular Virology, Ulm University Medical Center, 89081 Ulm, Germany
| | - Steffen Stenger
- Institute of Microbiology and Hygiene, Ulm University Medical Center, 89081 Ulm, Germany
| | - Christian Buske
- Institute of Experimental Cancer Research, Ulm University Medical Center, 89081 Ulm, Germany (M.K.)
| | - Palash Chandra Maity
- Institute of Experimental Cancer Research, Ulm University Medical Center, 89081 Ulm, Germany (M.K.)
| |
Collapse
|
3
|
Sun X, Wu Z, Su J, Li C. A deep attention model for wide-genome protein-peptide binding affinity prediction at a sequence level. Int J Biol Macromol 2024; 276:133811. [PMID: 38996881 DOI: 10.1016/j.ijbiomac.2024.133811] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2024] [Revised: 07/09/2024] [Accepted: 07/09/2024] [Indexed: 07/14/2024]
Abstract
Peptides are pivotal in numerous biological activities by engaging in up to 40 % of protein-protein interactions in many cellular processes. Due to their exceptional specificity and effectiveness, peptides have emerged as promising candidates for drug design. However, accurately predicting protein-peptide binding affinity remains a challenging. Aiming at the problem, we develop a prediction model PepPAP based on convolutional neural network and multi-head attention, which relies solely on sequence features. These features include physicochemical properties, intrinsic disorder, sequence encoding, and especially interface propensity which is extracted from 16,689 non-redundant protein-peptide complexes. Notably, the adopted regression stratification cross-validation scheme proposed in our previous work is beneficial to improve the prediction for the cases with extreme binding affinity values. On three benchmark test datasets: T100, a series of peptides targeting to PDZ domain and CXCR4, PepPAP shows excellent performance, outperforming the existing methods and demonstrating its good generalization ability. Furthermore, PepPAP has good results in binary interaction prediction, and the analysis of the feature space distribution visualization highlights PepPAP's effectiveness. To the best of our knowledge, PepPAP is the first sequence-based deep attention model for wide-genome protein-peptide binding affinity prediction, and holds the potential to offer valuable insights for the peptide-based drug design.
Collapse
Affiliation(s)
- Xiaohan Sun
- College of Chemistry and Life Science, Beijing University of Technology, Beijing 100124, China
| | - Zhixiang Wu
- College of Chemistry and Life Science, Beijing University of Technology, Beijing 100124, China
| | - Jingjie Su
- College of Chemistry and Life Science, Beijing University of Technology, Beijing 100124, China
| | - Chunhua Li
- College of Chemistry and Life Science, Beijing University of Technology, Beijing 100124, China.
| |
Collapse
|
4
|
Harms M, Haase A, Rodríguez-Alfonso A, Löffler J, Almeida-Hernández Y, Ruiz-Blanco YB, Albers D, Gilg A, von Bank F, Zech F, Groß R, Datta M, Jaikishan J, Draphoen B, Habib M, Ständker L, Wiese S, Lindén M, Winter G, Rasche V, Beer AJ, Jumaa H, Abadi AH, Kirchhoff F, Busch M, Dünker N, Sanchez-Garcia E, Münch J. Fatty acid conjugated EPI-X4 derivatives with increased activity and in vivo stability. J Control Release 2024; 373:583-598. [PMID: 39047872 DOI: 10.1016/j.jconrel.2024.07.049] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2024] [Revised: 07/09/2024] [Accepted: 07/21/2024] [Indexed: 07/27/2024]
Abstract
Dysregulation of the CXCL12/CXCR4 axis is implicated in autoimmune, inflammatory, and oncogenic diseases, positioning CXCR4 as a pivotal therapeutic target. We evaluated optimized variants of the specific endogenous CXCR4 antagonist, EPI-X4, addressing existing challenges in stability and potency. Our structure-activity relationship study investigates the conjugation of EPI-X4 derivatives with long-chain fatty acids, enhancing serum albumin interaction and receptor affinity. Molecular dynamic simulations revealed that the lipid moieties stabilize the peptide-receptor interaction through hydrophobic contacts at the receptor's N-terminus, anchoring the lipopeptide within the CXCR4 binding pocket and maintaining essential receptor interactions. Accordingly, lipidation resulted in increased receptor affinities and antagonistic activities. Additionally, by interacting with human serum albumin lipidated EPI-X4 derivatives displayed sustained stability in human plasma and extended circulation times in vivo. Selected candidates showed significant therapeutic potential in human retinoblastoma cells in vitro and in ovo, with our lead derivative exhibiting higher efficacies compared to its non-lipidated counterpart. This study not only elucidates the optimization trajectory for EPI-X4 derivatives but also underscores the intricate interplay between stability and efficacy, crucial for delineating their translational potential in clinical applications.
Collapse
Affiliation(s)
- Mirja Harms
- Institute of Molecular Virology, Ulm University Medical Center, Ulm 89081, Germany.
| | - André Haase
- Institute for Anatomy II, Department of Neuroanatomy, Center for Translational Neuro- and Behavioral Sciences (C-TNBS), Medical Faculty, University of Duisburg-Essen, 45147 Essen, Germany
| | - Armando Rodríguez-Alfonso
- Core Facility Functional Peptidomics, Ulm University Medical Center, Ulm 89081, Germany; Core Unit Mass Spectrometry and Proteomics, Ulm University Medical Center, Ulm 89081, Germany
| | - Jessica Löffler
- Department of Nuclear Medicine, Ulm University Medical Center, Ulm 89081, Germany
| | - Yasser Almeida-Hernández
- Computational Bioengineering, Department of Biochemical and Chemical Engineering, 44227 Dortmund, Germany
| | - Yasser B Ruiz-Blanco
- Computational Bioengineering, Department of Biochemical and Chemical Engineering, 44227 Dortmund, Germany
| | - Dan Albers
- Institute of Molecular Virology, Ulm University Medical Center, Ulm 89081, Germany
| | - Andrea Gilg
- Institute of Molecular Virology, Ulm University Medical Center, Ulm 89081, Germany
| | - Franziska von Bank
- Institute of Molecular Virology, Ulm University Medical Center, Ulm 89081, Germany
| | - Fabian Zech
- Institute of Molecular Virology, Ulm University Medical Center, Ulm 89081, Germany
| | - Rüdiger Groß
- Institute of Molecular Virology, Ulm University Medical Center, Ulm 89081, Germany
| | - Moumita Datta
- Institute of Immunology, Ulm University Medical Center, Ulm 89081, Germany
| | - Janeni Jaikishan
- Institute of Immunology, Ulm University Medical Center, Ulm 89081, Germany
| | | | - Monica Habib
- Department of Pharmaceutical Chemistry, Faculty of Pharmacy and Biotechnology, German University in Cairo, Cairo 11835, Egypt; Pharmaceutical Chemistry Department, School of Life and Medical Sciences, University of Hertfordshire Hosted by Global Academic Foundation, Cairo, Egypt
| | - Ludger Ständker
- Core Facility Functional Peptidomics, Ulm University Medical Center, Ulm 89081, Germany
| | - Sebastian Wiese
- Core Unit Mass Spectrometry and Proteomics, Ulm University Medical Center, Ulm 89081, Germany
| | - Mika Lindén
- Institute for Inorganic Chemistry II, 89081 Ulm, Germany
| | - Gordon Winter
- Department of Nuclear Medicine, Ulm University Medical Center, Ulm 89081, Germany
| | - Volker Rasche
- Experimental Cardiovascular Imaging (ExCaVI), Ulm University Medical Center, Ulm 89081, Germany
| | - Ambros J Beer
- Department of Nuclear Medicine, Ulm University Medical Center, Ulm 89081, Germany
| | - Hassan Jumaa
- Institute of Immunology, Ulm University Medical Center, Ulm 89081, Germany
| | - Ashraf H Abadi
- Department of Pharmaceutical Chemistry, Faculty of Pharmacy and Biotechnology, German University in Cairo, Cairo 11835, Egypt
| | - Frank Kirchhoff
- Institute of Molecular Virology, Ulm University Medical Center, Ulm 89081, Germany
| | - Maike Busch
- Institute for Anatomy II, Department of Neuroanatomy, Center for Translational Neuro- and Behavioral Sciences (C-TNBS), Medical Faculty, University of Duisburg-Essen, 45147 Essen, Germany
| | - Nicole Dünker
- Institute for Anatomy II, Department of Neuroanatomy, Center for Translational Neuro- and Behavioral Sciences (C-TNBS), Medical Faculty, University of Duisburg-Essen, 45147 Essen, Germany
| | - Elsa Sanchez-Garcia
- Computational Bioengineering, Department of Biochemical and Chemical Engineering, 44227 Dortmund, Germany
| | - Jan Münch
- Institute of Molecular Virology, Ulm University Medical Center, Ulm 89081, Germany
| |
Collapse
|
5
|
Pohl J, Litz A, El Ayoubi O, Rodríguez-Alfonso A, Ständker L, Harms M, Münch J, Jumaa H, Datta M. An Optimized Peptide Antagonist of CXCR4 Limits Survival of BCR-ABL1-Transformed Cells in Philadelphia-Chromosome-Positive B-Cell Acute Lymphoblastic Leukemia. Int J Mol Sci 2024; 25:8306. [PMID: 39125877 PMCID: PMC11312813 DOI: 10.3390/ijms25158306] [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: 05/16/2024] [Revised: 07/18/2024] [Accepted: 07/22/2024] [Indexed: 08/12/2024] Open
Abstract
Philadelphia-chromosome-positive acute lymphoblastic leukemia (Ph+ ALL) is characterized by reciprocal chromosomal translocation between chromosome 9 and 22, leading to the expression of constitutively active oncogenic BCR-ABL1 fusion protein. CXC chemokine receptor 4 (CXCR4) is essential for the survival of BCR-ABL1-transformed mouse pre-B cells, as the deletion of CXCR4 induces death in these cells. To investigate whether CXCR4 inhibition also effectively blocks BCR-ABL1-transformed cell growth in vitro, in this study, we explored an array of peptide-based inhibitors of CXCR4. The inhibitors were optimized derivatives of EPI-X4, an endogenous peptide antagonist of CXCR4. We observed that among all the candidates, EPI-X4 JM#170 (referred to as JM#170) effectively induced cell death in BCR-ABL1-transformed mouse B cells but had little effect on untransformed wild-type B cells. Importantly, AMD3100, a small molecule inhibitor of CXCR4, did not show this effect. Treatment with JM#170 induced transient JNK phosphorylation in BCR-ABL1-transformed cells, which in turn activated the intrinsic apoptotic pathway by inducing cJun, Bim, and Bax gene expressions. Combinatorial treatment of JM#170 with ABL1 kinase inhibitor Imatinib exerted a stronger killing effect on BCR-ABL1-transformed cells even at a lower dose of Imatinib. Surprisingly, JM#170 actively killed Sup-B15 cells, a BCR-ABL1+ human ALL cell line, but had no effect on the BCR-ABL1- 697 cell line. This suggests that the inhibitory effect of JM#170 is specific for BCR-ABL1+ ALL. Taken together, JM#170 emerges as a potent novel drug against Ph+ ALL.
Collapse
Affiliation(s)
- Johanna Pohl
- Institute of Immunology, Ulm University Medical Center, 89081 Ulm, Germany (O.E.A.)
| | - Angela Litz
- Institute of Immunology, Ulm University Medical Center, 89081 Ulm, Germany (O.E.A.)
| | - Omar El Ayoubi
- Institute of Immunology, Ulm University Medical Center, 89081 Ulm, Germany (O.E.A.)
| | - Armando Rodríguez-Alfonso
- Core Facility Functional Peptidomics, Ulm University Medical Center, 89081 Ulm, Germany; (A.R.-A.); (L.S.)
- Core Unit Mass Spectrometry and Proteomics, Ulm University Medical Center, 89081 Ulm, Germany
| | - Ludger Ständker
- Core Facility Functional Peptidomics, Ulm University Medical Center, 89081 Ulm, Germany; (A.R.-A.); (L.S.)
| | - Mirja Harms
- Institute of Molecular Virology, Ulm University Medical Center, 89081 Ulm, Germany; (M.H.); (J.M.)
| | - Jan Münch
- Institute of Molecular Virology, Ulm University Medical Center, 89081 Ulm, Germany; (M.H.); (J.M.)
| | - Hassan Jumaa
- Institute of Immunology, Ulm University Medical Center, 89081 Ulm, Germany (O.E.A.)
| | - Moumita Datta
- Institute of Immunology, Ulm University Medical Center, 89081 Ulm, Germany (O.E.A.)
| |
Collapse
|
6
|
Zuo Q, Li Y, Lai X, Bao G, Chen L, He Z, Song X, E R, Wang P, Shi Y, Luo H, Sun W, Wang R. Cysteine-Specific Multifaceted Bioconjugation of Peptides and Proteins Using 5-Substituted 1,2,3-Triazines. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2024; 11:e2308491. [PMID: 38466927 DOI: 10.1002/advs.202308491] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/07/2023] [Revised: 02/08/2024] [Indexed: 03/13/2024]
Abstract
Peptide and protein postmodification have gained significant attention due to their extensive impact on biomolecule engineering and drug discovery, of which cysteine-specific modification strategies are prominent due to their inherent nucleophilicity and low abundance. Herein, the study introduces a novel approach utilizing multifunctional 5-substituted 1,2,3-triazine derivatives to achieve multifaceted bioconjugation targeting cysteine-containing peptides and proteins. On the one hand, this represents an inaugural instance of employing 1,2,3-triazine in biomolecular-specific modification within a physiological solution. On the other hand, as a powerful combination of precision modification and biorthogonality, this strategy allows for the one-pot dual-orthogonal functionalization of biomolecules utilizing the aldehyde group generated simultaneously. 1,2,3-Triazine derivatives with diverse functional groups allow conjugation to peptides or proteins, while bi-triazines enable peptide cyclization and dimerization. The examination of the stability of bi-triazines revealed their potential for reversible peptide modification. This work establishes a comprehensive platform for identifying cysteine-selective modifications, providing new avenues for peptide-based drug development, protein bioconjugation, and chemical biology research.
Collapse
Affiliation(s)
- Quan Zuo
- State Key Laboratory of Bioactive Substance and Function of Natural Medicines, Institute of Materia Medica, Chinese Academy of Medical Sciences and Peking Union Medical College, Xian Nong Tan Street, Beijing, 100050, P. R. China
| | - Yiping Li
- Key Laboratory of Preclinical Study for New Drugs of Gansu Province, School of Basic Medical Sciences and Research Unit of Peptide Science, Chinese Academy of Medical Sciences, Lanzhou University, 199 West Donggang Road, Lanzhou, Gansu, 730000, P. R. China
| | - Xuanliang Lai
- State Key Laboratory of Bioactive Substance and Function of Natural Medicines, Institute of Materia Medica, Chinese Academy of Medical Sciences and Peking Union Medical College, Xian Nong Tan Street, Beijing, 100050, P. R. China
| | - Guangjun Bao
- Key Laboratory of Preclinical Study for New Drugs of Gansu Province, School of Basic Medical Sciences and Research Unit of Peptide Science, Chinese Academy of Medical Sciences, Lanzhou University, 199 West Donggang Road, Lanzhou, Gansu, 730000, P. R. China
| | - Lu Chen
- State Key Laboratory of Bioactive Substance and Function of Natural Medicines, Institute of Materia Medica, Chinese Academy of Medical Sciences and Peking Union Medical College, Xian Nong Tan Street, Beijing, 100050, P. R. China
| | - Zeyuan He
- Key Laboratory of Preclinical Study for New Drugs of Gansu Province, School of Basic Medical Sciences and Research Unit of Peptide Science, Chinese Academy of Medical Sciences, Lanzhou University, 199 West Donggang Road, Lanzhou, Gansu, 730000, P. R. China
| | - Xinyi Song
- Key Laboratory of Preclinical Study for New Drugs of Gansu Province, School of Basic Medical Sciences and Research Unit of Peptide Science, Chinese Academy of Medical Sciences, Lanzhou University, 199 West Donggang Road, Lanzhou, Gansu, 730000, P. R. China
| | - Ruiyao E
- Key Laboratory of Preclinical Study for New Drugs of Gansu Province, School of Basic Medical Sciences and Research Unit of Peptide Science, Chinese Academy of Medical Sciences, Lanzhou University, 199 West Donggang Road, Lanzhou, Gansu, 730000, P. R. China
| | - Pengxin Wang
- State Key Laboratory of Bioactive Substance and Function of Natural Medicines, Institute of Materia Medica, Chinese Academy of Medical Sciences and Peking Union Medical College, Xian Nong Tan Street, Beijing, 100050, P. R. China
| | - Yuntao Shi
- State Key Laboratory of Bioactive Substance and Function of Natural Medicines, Institute of Materia Medica, Chinese Academy of Medical Sciences and Peking Union Medical College, Xian Nong Tan Street, Beijing, 100050, P. R. China
| | - Huixin Luo
- State Key Laboratory of Bioactive Substance and Function of Natural Medicines, Institute of Materia Medica, Chinese Academy of Medical Sciences and Peking Union Medical College, Xian Nong Tan Street, Beijing, 100050, P. R. China
| | - Wangsheng Sun
- Key Laboratory of Preclinical Study for New Drugs of Gansu Province, School of Basic Medical Sciences and Research Unit of Peptide Science, Chinese Academy of Medical Sciences, Lanzhou University, 199 West Donggang Road, Lanzhou, Gansu, 730000, P. R. China
| | - Rui Wang
- State Key Laboratory of Bioactive Substance and Function of Natural Medicines, Institute of Materia Medica, Chinese Academy of Medical Sciences and Peking Union Medical College, Xian Nong Tan Street, Beijing, 100050, P. R. China
- Key Laboratory of Preclinical Study for New Drugs of Gansu Province, School of Basic Medical Sciences and Research Unit of Peptide Science, Chinese Academy of Medical Sciences, Lanzhou University, 199 West Donggang Road, Lanzhou, Gansu, 730000, P. R. China
| |
Collapse
|
7
|
Sagini MN, Zepp M, Eyol E, Ali DM, Gromova S, Dahlmann M, Behrens D, Groeschel C, Tischmeier L, Hoffmann J, Berger MR, Forssmann WG. EPI-X4, a CXCR4 antagonist inhibits tumor growth in pancreatic cancer and lymphoma models. Peptides 2024; 175:171111. [PMID: 38036098 DOI: 10.1016/j.peptides.2023.171111] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/06/2023] [Revised: 10/20/2023] [Accepted: 10/25/2023] [Indexed: 12/02/2023]
Abstract
Endogenous peptide inhibitor for CXCR4 (EPI-X4) is a CXCR4 antagonist with potential for cancer therapy. It is a processed fragment of serum albumin from the hemofiltrate of dialysis patients. This study reports the efficacy of fifteen EPI-X4 derivatives in pancreatic cancer and lymphoma models. In vitro, the peptides were investigated for antiproliferation (cytotoxicity) by MTT assay. The mRNA expression for CXCR4 and CXCL12 was determined by RT-PCR, chip array and RNA sequencing. Chip array analysis yielded 634 genes associated with CXCR4/CXCL12 signaling. About 21% of these genes correlated with metastasis in the context of cell motility, proliferation, and survival. Expression levels of these genes were altered in pancreatic cancer (36%), lymphoma models (53%) and in patients' data (58%). EPI-X4 derivatives failed to inhibit cell proliferation due to low expression of CXCR4 in vitro, but inhibited tumor growth in the bioassays with significant efficacy. In the pancreatic cancer model, EPI-X4a, f and k inhibited mean tumor growth by > 50% and even caused complete remissions. In the lymphoma model, EPI-X4b, n and p inhibited mean tumor growth by > 70% and caused stable disease. Given the non-toxic and non-immunogenic properties of EPI-X4, these findings underscore its status as a promising therapy of pancreatic cancer and lymphoma and warrant further studies. SIMPLE SUMMARY: This study examined the value of chemokine receptor CXCR4 as an antineoplastic target for the endogenous peptide inhibitor of CXCR4 (EPI-X4), a 12-meric peptide derived from serum albumin. EPI-X4 inhibits CXCR4 interaction with its natural ligand, CXCL12 (SDF1). Therefore, malignancies (including pancreatic cancer and lymphoma) that depend on the CXCR4/CXCL12 pathway for progression can be targeted with EPI-X4. Of 634 genes that were linked to the CXCR4/CXCL12 pathway, 21% were associated with metastasis. In cultured human Suit2-007 pancreatic cancer cells, CXCR4 showed low to undetectable expression, which was why EPI-X4 did not inhibit pancreatic cancer cell proliferation. These findings were different in vivo, where CXCR4 was highly expressed and EPI-X4 inhibited tumor growth in rodents harboring pancreatic cancer or lymphoma. In the pancreatic cancer model, EPI-X4 derivatives a, f and k caused complete remissions, while in lymphomas EPI-X4 derivatives b, n and p caused stable disease.
Collapse
Affiliation(s)
- Micah N Sagini
- Toxicology and Chemotherapy Unit, German Cancer Research Centre (DKFZ), Heidelberg, Germany
| | - Michael Zepp
- Toxicology and Chemotherapy Unit, German Cancer Research Centre (DKFZ), Heidelberg, Germany
| | - Ergül Eyol
- Toxicology and Chemotherapy Unit, German Cancer Research Centre (DKFZ), Heidelberg, Germany
| | - Doaa M Ali
- Toxicology and Chemotherapy Unit, German Cancer Research Centre (DKFZ), Heidelberg, Germany
| | - Svetlana Gromova
- EPO, Experimental Pharmacology & Oncology Berlin-Buch GmbH, Germany
| | - Mathias Dahlmann
- EPO, Experimental Pharmacology & Oncology Berlin-Buch GmbH, Germany
| | - Diana Behrens
- EPO, Experimental Pharmacology & Oncology Berlin-Buch GmbH, Germany
| | - Christian Groeschel
- NeoPep Pharma GmbH & Co. KG., Hannover, Germany and Hannover Medical School, Department of Internal Medicine, Germany
| | - Linus Tischmeier
- NeoPep Pharma GmbH & Co. KG., Hannover, Germany and Hannover Medical School, Department of Internal Medicine, Germany
| | - Jens Hoffmann
- EPO, Experimental Pharmacology & Oncology Berlin-Buch GmbH, Germany
| | - Martin R Berger
- Toxicology and Chemotherapy Unit, German Cancer Research Centre (DKFZ), Heidelberg, Germany.
| | - Wolf-Georg Forssmann
- NeoPep Pharma GmbH & Co. KG., Hannover, Germany and Hannover Medical School, Department of Internal Medicine, Germany.
| |
Collapse
|
8
|
White CW, Platt S, Kilpatrick LE, Dale N, Abhayawardana RS, Dekkers S, Kindon ND, Kellam B, Stocks MJ, Pfleger KDG, Hill SJ. CXCL17 is an allosteric inhibitor of CXCR4 through a mechanism of action involving glycosaminoglycans. Sci Signal 2024; 17:eabl3758. [PMID: 38502733 PMCID: PMC7615768 DOI: 10.1126/scisignal.abl3758] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2021] [Accepted: 02/29/2024] [Indexed: 03/21/2024]
Abstract
CXCL17 is a chemokine principally expressed by mucosal tissues, where it facilitates chemotaxis of monocytes, dendritic cells, and macrophages and has antimicrobial properties. CXCL17 is also implicated in the pathology of inflammatory disorders and progression of several cancers, and its expression is increased during viral infections of the lung. However, the exact role of CXCL17 in health and disease requires further investigation, and there is a need for confirmed molecular targets mediating CXCL17 functional responses. Using a range of bioluminescence resonance energy transfer (BRET)-based assays, here we demonstrated that CXCL17 inhibited CXCR4-mediated signaling and ligand binding. Moreover, CXCL17 interacted with neuropillin-1, a VEGFR2 coreceptor. In addition, we found that CXCL17 only inhibited CXCR4 ligand binding in intact cells and demonstrated that this effect was mimicked by known glycosaminoglycan binders, surfen and protamine sulfate. Disruption of putative GAG binding domains in CXCL17 prevented CXCR4 binding. This indicated that CXCL17 inhibited CXCR4 by a mechanism of action that potentially required the presence of a glycosaminoglycan-containing accessory protein. Together, our results revealed that CXCL17 is an endogenous inhibitor of CXCR4 and represents the next step in our understanding of the function of CXCL17 and regulation of CXCR4 signaling.
Collapse
Affiliation(s)
- Carl W. White
- Cell Signalling and Pharmacology Research Group, Division of Physiology, Pharmacology & Neuroscience, School of Life Sciences, University of Nottingham, Nottingham NG7 2UH, United Kingdom
- Centre of Membrane Proteins and Receptors, University of Birmingham and University of Nottingham, The Midlands, UK
- Harry Perkins Institute of Medical Research and Centre for Medical Research, The University of Western Australia, QEII Medical Centre, Nedlands, Western Australia 6009, Australia
- Australian Research Council Centre for Personalised Therapeutics Technologies, Australia
- Dimerix Limited, Melbourne, Australia
| | - Simon Platt
- Cell Signalling and Pharmacology Research Group, Division of Physiology, Pharmacology & Neuroscience, School of Life Sciences, University of Nottingham, Nottingham NG7 2UH, United Kingdom
- Centre of Membrane Proteins and Receptors, University of Birmingham and University of Nottingham, The Midlands, UK
| | - Laura E. Kilpatrick
- Centre of Membrane Proteins and Receptors, University of Birmingham and University of Nottingham, The Midlands, UK
- School of Pharmacy, Biodiscovery Institute, University of Nottingham, Nottingham NG7 2RD, United Kingdom
| | - Natasha Dale
- Harry Perkins Institute of Medical Research and Centre for Medical Research, The University of Western Australia, QEII Medical Centre, Nedlands, Western Australia 6009, Australia
- Australian Research Council Centre for Personalised Therapeutics Technologies, Australia
| | - Rekhati S. Abhayawardana
- Harry Perkins Institute of Medical Research and Centre for Medical Research, The University of Western Australia, QEII Medical Centre, Nedlands, Western Australia 6009, Australia
- Australian Research Council Centre for Personalised Therapeutics Technologies, Australia
| | - Sebastian Dekkers
- Cell Signalling and Pharmacology Research Group, Division of Physiology, Pharmacology & Neuroscience, School of Life Sciences, University of Nottingham, Nottingham NG7 2UH, United Kingdom
- Centre of Membrane Proteins and Receptors, University of Birmingham and University of Nottingham, The Midlands, UK
- School of Pharmacy, Biodiscovery Institute, University of Nottingham, Nottingham NG7 2RD, United Kingdom
| | - Nicholas D Kindon
- Centre of Membrane Proteins and Receptors, University of Birmingham and University of Nottingham, The Midlands, UK
- School of Pharmacy, Biodiscovery Institute, University of Nottingham, Nottingham NG7 2RD, United Kingdom
| | - Barrie Kellam
- Centre of Membrane Proteins and Receptors, University of Birmingham and University of Nottingham, The Midlands, UK
- School of Pharmacy, Biodiscovery Institute, University of Nottingham, Nottingham NG7 2RD, United Kingdom
| | - Michael J Stocks
- School of Pharmacy, Biodiscovery Institute, University of Nottingham, Nottingham NG7 2RD, United Kingdom
| | - Kevin D. G. Pfleger
- Harry Perkins Institute of Medical Research and Centre for Medical Research, The University of Western Australia, QEII Medical Centre, Nedlands, Western Australia 6009, Australia
- Australian Research Council Centre for Personalised Therapeutics Technologies, Australia
- Dimerix Limited, Melbourne, Australia
| | - Stephen J. Hill
- Cell Signalling and Pharmacology Research Group, Division of Physiology, Pharmacology & Neuroscience, School of Life Sciences, University of Nottingham, Nottingham NG7 2UH, United Kingdom
- Centre of Membrane Proteins and Receptors, University of Birmingham and University of Nottingham, The Midlands, UK
| |
Collapse
|
9
|
Schrader M. Origins, Technological Advancement, and Applications of Peptidomics. Methods Mol Biol 2024; 2758:3-47. [PMID: 38549006 DOI: 10.1007/978-1-0716-3646-6_1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/02/2024]
Abstract
Peptidomics is the comprehensive characterization of peptides from biological sources instead of heading for a few single peptides in former peptide research. Mass spectrometry allows to detect a multitude of peptides in complex mixtures and thus enables new strategies leading to peptidomics. The term was established in the year 2001, and up to now, this new field has grown to over 3000 publications. Analytical techniques originally developed for fast and comprehensive analysis of peptides in proteomics were specifically adjusted for peptidomics. Although it is thus closely linked to proteomics, there are fundamental differences with conventional bottom-up proteomics. Fundamental technological advancements of peptidomics since have occurred in mass spectrometry and data processing, including quantification, and more slightly in separation technology. Different strategies and diverse sources of peptidomes are mentioned by numerous applications, such as discovery of neuropeptides and other bioactive peptides, including the use of biochemical assays. Furthermore, food and plant peptidomics are introduced similarly. Additionally, applications with a clinical focus are included, comprising biomarker discovery as well as immunopeptidomics. This overview extensively reviews recent methods, strategies, and applications including links to all other chapters of this book.
Collapse
Affiliation(s)
- Michael Schrader
- Department of Bioengineering Sciences, Weihenstephan-Tr. University of Applied Sciences, Freising, Germany.
| |
Collapse
|
10
|
Harms M, Fabech Hansson R, Gilg A, Almeida-Hernández Y, Löffler J, Rodríguez-Alfonso A, Habib MMW, Albers D, Ahmed NS, Abadi AH, Winter G, Rasche V, Beer AJ, Weidinger G, Preising N, Ständker L, Wiese S, Sanchez-Garcia E, Zelikin AN, Münch J. Development of N-Terminally Modified Variants of the CXCR4-Antagonistic Peptide EPI-X4 for Enhanced Plasma Stability. J Med Chem 2023; 66:15189-15204. [PMID: 37940118 PMCID: PMC10682998 DOI: 10.1021/acs.jmedchem.3c01128] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2023] [Indexed: 11/10/2023]
Abstract
EPI-X4, a natural peptide CXCR4 antagonist, shows potential for treating inflammation and cancer, but its short plasma stability limits its clinical application. We aimed to improve the plasma stability of EPI-X4 analogues without compromising CXCR4 antagonism. Our findings revealed that only the peptide N-terminus is prone to degradation. Consequently, incorporating d-amino acids or acetyl groups in this region enhanced peptide stability in plasma. Notably, EPI-X4 leads 5, 27, and 28 not only retained their CXCR4 binding and antagonism but also remained stable in plasma for over 8 h. Molecular dynamic simulations showed that these modified analogues bind similarly to CXCR4 as the original peptide. To further increase their systemic half-lives, we conjugated these stabilized analogues with large polymers and albumin binders. These advances highlight the potential of the optimized EPI-X4 analogues as promising CXCR4-targeted therapeutics and set the stage for more detailed preclinical assessments.
Collapse
Affiliation(s)
- Mirja Harms
- Institute
of Molecular Virology, Ulm University Medical
Center, Ulm 89081, Germany
| | - Rikke Fabech Hansson
- Department
of Chemistry and iNANO Interdisciplinary Nanoscience Centre, Aarhus University, Aarhus 8000, Denmark
| | - Andrea Gilg
- Institute
of Molecular Virology, Ulm University Medical
Center, Ulm 89081, Germany
| | - Yasser Almeida-Hernández
- Department
of Biochemical and Chemical Engineering, Computational Bioengineering, Emil-Figge Str. 66, Dortmund 44227, Germany
| | - Jessica Löffler
- Department
of Nuclear Medicine, Ulm University Medical
Center, Ulm 89081, Germany
| | - Armando Rodríguez-Alfonso
- Core
Facility Functional Peptidomics, Ulm University
Medical Center, Ulm 89081, Germany
- Core Unit
Mass Spectrometry and Proteomics, Ulm University
Medical Center, Ulm 89081, Germany
| | - Monica M. W. Habib
- Department
of Pharmaceutical Chemistry, Faculty of Pharmacy and Biotechnology, German University in Cairo, Cairo 11835, Egypt
- Pharmaceutical
Chemistry Department, School of Life and Medical Sciences, University of Hertfordshire Hosted by Global Academic
Foundation, Cairo 11865, Egypt
| | - Dan Albers
- Institute
of Molecular Virology, Ulm University Medical
Center, Ulm 89081, Germany
| | - Nermin S. Ahmed
- Department
of Pharmaceutical Chemistry, Faculty of Pharmacy and Biotechnology, German University in Cairo, Cairo 11835, Egypt
| | - Ashraf H. Abadi
- Department
of Pharmaceutical Chemistry, Faculty of Pharmacy and Biotechnology, German University in Cairo, Cairo 11835, Egypt
| | - Gordon Winter
- Department
of Nuclear Medicine, Ulm University Medical
Center, Ulm 89081, Germany
| | - Volker Rasche
- Experimental
Cardiovascular Imaging (ExCaVI), Ulm University
Medical Center, Ulm 89081, Germany
| | - Ambros J. Beer
- Department
of Nuclear Medicine, Ulm University Medical
Center, Ulm 89081, Germany
| | - Gilbert Weidinger
- Institute
of Biochemistry and Molecular Biology, Ulm
University, Ulm 89081, Germany
| | - Nico Preising
- Core
Facility Functional Peptidomics, Ulm University
Medical Center, Ulm 89081, Germany
| | - Ludger Ständker
- Core
Facility Functional Peptidomics, Ulm University
Medical Center, Ulm 89081, Germany
| | - Sebastian Wiese
- Core Unit
Mass Spectrometry and Proteomics, Ulm University
Medical Center, Ulm 89081, Germany
| | - Elsa Sanchez-Garcia
- Department
of Biochemical and Chemical Engineering, Computational Bioengineering, Emil-Figge Str. 66, Dortmund 44227, Germany
| | - Alexander N. Zelikin
- Department
of Chemistry and iNANO Interdisciplinary Nanoscience Centre, Aarhus University, Aarhus 8000, Denmark
| | - Jan Münch
- Institute
of Molecular Virology, Ulm University Medical
Center, Ulm 89081, Germany
- Core
Facility Functional Peptidomics, Ulm University
Medical Center, Ulm 89081, Germany
| |
Collapse
|
11
|
Jung CK, Münch J, Jacob T. Conformational States of the CXCR4 Inhibitor Peptide EPI-X4-A Theoretical Analysis. Int J Mol Sci 2023; 24:16229. [PMID: 38003419 PMCID: PMC10671355 DOI: 10.3390/ijms242216229] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2023] [Revised: 10/30/2023] [Accepted: 11/02/2023] [Indexed: 11/26/2023] Open
Abstract
EPI-X4, an endogenous peptide inhibitor, has exhibited potential as a blocker of CXCR4-a G protein-coupled receptor. This unique inhibitor demonstrates the ability to impede HIV-1 infection and halt CXCR4-dependent processes such as tumor cell migration and invagination. Despite its promising effects, a comprehensive understanding of the interaction between EPI-X4 and CXCR4 under natural conditions remains elusive due to experimental limitations. To bridge this knowledge gap, a simulation approach was undertaken. Approximately 150,000 secondary structures of EPI-X4 were subjected to simulations to identify thermodynamically stable candidates. This simulation process harnessed a self-developed reactive force field operating within the ReaxFF framework. The application of the Two-Phase Thermodynamic methodology to ReaxFF facilitated the derivation of crucial thermodynamic attributes of the EPI-X4 conformers. To deepen insights, an ab initio density functional theory calculation method was employed to assess the electrostatic potentials of the most relevant (i.e., stable) EPI-X4 structures. This analytical endeavor aimed to enhance comprehension of the inhibitor's structural characteristics. As a result of these investigations, predictions were made regarding how EPI-X4 interacts with CXCR4. Two pivotal requirements emerged. Firstly, the spatial conformation of EPI-X4 must align effectively with the CXCR4 receptor protein. Secondly, the functional groups present on the surface of the inhibitor's structure must complement the corresponding features of CXCR4 to induce attraction between the two entities. These predictive outcomes were based on a meticulous analysis of the conformers, conducted in a gaseous environment. Ultimately, this rigorous exploration yielded a suitable EPI-X4 structure that fulfills the spatial and functional prerequisites for interacting with CXCR4, thus potentially shedding light on new avenues for therapeutic development.
Collapse
Affiliation(s)
- Christoph Karsten Jung
- Helmholtz Institute Ulm (HIU) Electrochemical Energy Storage, Helmholtzstr. 11, D-89081 Ulm, Germany
- Karlsruhe Institute of Technology (KIT), D-76021 Karlsruhe, Germany
- Institute of Electrochemistry, Ulm University, Albert-Einstein-Allee 47, D-89081 Ulm, Germany
| | - Jan Münch
- Institute of Molecular Virology, Ulm University Medical Center, Meyerhofstr. 1, D-89081 Ulm, Germany
| | - Timo Jacob
- Helmholtz Institute Ulm (HIU) Electrochemical Energy Storage, Helmholtzstr. 11, D-89081 Ulm, Germany
- Karlsruhe Institute of Technology (KIT), D-76021 Karlsruhe, Germany
- Institute of Electrochemistry, Ulm University, Albert-Einstein-Allee 47, D-89081 Ulm, Germany
| |
Collapse
|
12
|
Schauenburg D, Zech F, Heck AJ, von Maltitz P, Harms M, Führer S, Alleva N, Münch J, Kuan SL, Kirchhoff F, Weil T. Peptide Bispecifics Inhibiting HIV-1 Infection by an Orthogonal Chemical and Supramolecular Strategy. Bioconjug Chem 2023; 34:1645-1652. [PMID: 37665137 PMCID: PMC10515486 DOI: 10.1021/acs.bioconjchem.3c00314] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 09/05/2023]
Abstract
Viral infections pose a significant threat to human health, and effective antiviral strategies are urgently needed. Antiviral peptides have emerged as a promising class of therapeutic agents due to their unique properties and mechanisms of action. While effective on their own, combining antiviral peptides may allow us to enhance their potency and to prevent viral resistance. Here, we developed an orthogonal chemical strategy to prepare a heterodimeric peptide conjugate assembled on a protein-based nanoplatform. Specifically, we combined the optimized version of two peptides inhibiting HIV-1 by distinct mechanisms. Virus-inhibitory peptide (VIRIP) is a 20 amino acid fragment of α1-antitrypsin that inhibits HIV-1 by targeting the gp41 fusion peptide. Endogenous peptide inhibitor of CXCR4 (EPI-X4) is a 16-residue fragment of human serum albumin that prevents HIV-1 entry by binding to the viral CXCR4 co-receptor. Optimized forms of both peptides are assembled on supramolecular nanoplatforms through the streptavidin-biotin interaction. We show that the construct consisting of the two different peptides (SAv-VIR-102C9-EPI-X4 JM#173-C) shows increased activity against CCR5- and CXCR4-tropic HIV-1 variants. Our results are a proof of concept that peptides with different modes of action can be assembled on nanoplatforms to enhance their antiviral activity.
Collapse
Affiliation(s)
- Dominik Schauenburg
- Max-Planck Institute for Polymer Research, Ackermannweg 10, 55128 Mainz, Germany
| | - Fabian Zech
- Institute of Molecular Virology, Ulm University Medical Center, Meyerhofstr. 1, 89081 Ulm, Germany
| | - Astrid Johanna Heck
- Max-Planck Institute for Polymer Research, Ackermannweg 10, 55128 Mainz, Germany
| | - Pascal von Maltitz
- Institute of Molecular Virology, Ulm University Medical Center, Meyerhofstr. 1, 89081 Ulm, Germany
| | - Mirja Harms
- Institute of Molecular Virology, Ulm University Medical Center, Meyerhofstr. 1, 89081 Ulm, Germany
| | - Siska Führer
- Max-Planck Institute for Polymer Research, Ackermannweg 10, 55128 Mainz, Germany
| | - Nico Alleva
- Max-Planck Institute for Polymer Research, Ackermannweg 10, 55128 Mainz, Germany
| | - Jan Münch
- Institute of Molecular Virology, Ulm University Medical Center, Meyerhofstr. 1, 89081 Ulm, Germany
| | - Seah Ling Kuan
- Max-Planck Institute for Polymer Research, Ackermannweg 10, 55128 Mainz, Germany
| | - Frank Kirchhoff
- Institute of Molecular Virology, Ulm University Medical Center, Meyerhofstr. 1, 89081 Ulm, Germany
| | - Tanja Weil
- Max-Planck Institute for Polymer Research, Ackermannweg 10, 55128 Mainz, Germany
| |
Collapse
|
13
|
Bao S, Darvishi M, H Amin A, Al-Haideri MT, Patra I, Kashikova K, Ahmad I, Alsaikhan F, Al-Qaim ZH, Al-Gazally ME, Kiasari BA, Tavakoli-Far B, Sidikov AA, Mustafa YF, Akhavan-Sigari R. CXC chemokine receptor 4 (CXCR4) blockade in cancer treatment. J Cancer Res Clin Oncol 2023; 149:7945-7968. [PMID: 36905421 DOI: 10.1007/s00432-022-04444-w] [Citation(s) in RCA: 14] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2022] [Accepted: 10/19/2022] [Indexed: 03/12/2023]
Abstract
CXC chemokine receptor type 4 (CXCR4) is a member of the G protein-coupled receptors (GPCRs) superfamily and is specific for CXC chemokine ligand 12 (CXCL12, also known as SDF-1), which makes CXCL12/CXCR4 axis. CXCR4 interacts with its ligand, triggering downstream signaling pathways that influence cell proliferation chemotaxis, migration, and gene expression. The interaction also regulates physiological processes, including hematopoiesis, organogenesis, and tissue repair. Multiple evidence revealed that CXCL12/CXCR4 axis is implicated in several pathways involved in carcinogenesis and plays a key role in tumor growth, survival, angiogenesis, metastasis, and therapeutic resistance. Several CXCR4-targeting compounds have been discovered and used for preclinical and clinical cancer therapy, most of which have shown promising anti-tumor activity. In this review, we summarized the physiological signaling of the CXCL12/CXCR4 axis and described the role of this axis in tumor progression, and focused on the potential therapeutic options and strategies to block CXCR4.
Collapse
Affiliation(s)
- Shunshun Bao
- The First Clinical Medical College, Xuzhou Medical University, 221000, Xuzhou, China
| | - Mohammad Darvishi
- Infectious Diseases and Tropical Medicine Research Center (IDTMRC), Department of Aerospace and Subaquatic Medicine, AJA University of Medicinal Sciences, Tehran, Iran
| | - Ali H Amin
- Deanship of Scientific Research, Umm Al-Qura University, 21955, Makkah, Saudi Arabia
- Zoology Department, Faculty of Science, Mansoura University, 35516, Mansoura, Egypt
| | - Maysoon T Al-Haideri
- Department of Physiotherapy, Cihan University-Erbil, Erbil, Kurdistan Region, Iraq
| | - Indrajit Patra
- An Independent Researcher, National Institute of Technology Durgapur, Durgapur, West Bengal, India
| | | | - Irfan Ahmad
- Department of Clinical Laboratory Sciences, College of Applied Medical Sciences, King Khalid University, Abha, Saudi Arabia
| | - Fahad Alsaikhan
- College of Pharmacy, Prince Sattam Bin Abdulaziz University, Alkharj, Saudi Arabia
| | | | | | - Bahman Abedi Kiasari
- Virology Department, Faculty of Veterinary Medicine, The University of Tehran, Tehran, Iran.
| | - Bahareh Tavakoli-Far
- Dietary Supplements and Probiotic Research Center, Alborz University of Medical Sciences, Karaj, Iran.
- Department of Physiology and Pharmacology, Faculty of Medicine, Alborz University of Medical Sciences, Karaj, Iran.
| | - Akmal A Sidikov
- Rector, Ferghana Medical Institute of Public Health, Ferghana, Uzbekistan
| | - Yasser Fakri Mustafa
- Department of Pharmaceutical Chemistry, College of Pharmacy, University of Mosul, Mosul, 41001, Iraq
| | - Reza Akhavan-Sigari
- Department of Neurosurgery, University Medical Center Tuebingen, Tübingen, Germany
- Department of Health Care Management and Clinical Research, Collegium Humanum Warsaw Management University, Warsaw, Poland
| |
Collapse
|
14
|
Gaonkar RH, Schmidt YT, Mansi R, Almeida-Hernanadez Y, Sanchez-Garcia E, Harms M, Münch J, Fani M. Development of a New Class of CXCR4-Targeting Radioligands Based on the Endogenous Antagonist EPI-X4 for Oncological Applications. J Med Chem 2023. [PMID: 37328158 DOI: 10.1021/acs.jmedchem.3c00131] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/18/2023]
Abstract
The peptide fragment of human serum albumin that was identified as an inhibitor of C-X-C motif chemokine receptor 4 (CXCR4), termed EPI-X4, was investigated as a scaffold for the development of CXCR4-targeting radio-theragnostics. Derivatives of its truncated version JM#21 (ILRWSRKLPCVS) were conjugated to 1,4,7,10-tetraazacyclododecane-1,4,7,10-tetraacetic acid (DOTA) and tested in Jurkat and Ghost-CXCR4 cells. Ligand-1, -2, -5, -6, -7, -8, and -9 were selected for radiolabeling. Molecular modeling indicated that 177Lu-DOTA incorporation C-terminally did not interfere with the CXCR4 binding. Lipophilicity, in vitro plasma stability, and cellular uptake hinted 177Lu-7 as superior. In Jurkat xenografts, all radioligands showed >90% washout from the body within an hour, with the exception of 177Lu-7 and 177Lu-9. 177Lu-7 demonstrated best CXCR4-tumor targeting. Ex vivo biodistribution and single-photon emission computed tomography (SPECT)/positron emission tomography (PET)/CT imaging of 177Lu-7/68Ga-7 showed the same distribution profile for both radioligands, characterized by very low uptake in all nontargeted organs except the kidneys. The data support the feasibility of CXCR4-targeting with EPI-X4-based radioligands and designate ligand-7 as a lead candidate for further optimization.
Collapse
Affiliation(s)
- Raghuvir Haridas Gaonkar
- Division of Radiopharmaceutical Chemistry, Department Theragnostics, University Hospital Basel, Basel 4031, Switzerland
| | - Yannik Tim Schmidt
- Division of Radiopharmaceutical Chemistry, Department Theragnostics, University Hospital Basel, Basel 4031, Switzerland
| | - Rosalba Mansi
- Division of Radiopharmaceutical Chemistry, Department Theragnostics, University Hospital Basel, Basel 4031, Switzerland
| | - Yasser Almeida-Hernanadez
- Computational Biochemistry, Center of Medical Biotechnology, University of Duisburg-Essen, Essen 45117, Germany
- Computational Bioengineering, Faculty of Bio- and Chemical Engineering, Technical University Dortmund, Dortmund 44227, Germany
| | - Elsa Sanchez-Garcia
- Computational Biochemistry, Center of Medical Biotechnology, University of Duisburg-Essen, Essen 45117, Germany
- Computational Bioengineering, Faculty of Bio- and Chemical Engineering, Technical University Dortmund, Dortmund 44227, Germany
| | - Mirja Harms
- Institute of Molecular Virology, Ulm University Medical Center, Ulm 89081, Germany
| | - Jan Münch
- Institute of Molecular Virology, Ulm University Medical Center, Ulm 89081, Germany
- Core Facility Functional Peptidomics, Ulm University Medical Center, Ulm 89081, Germany
| | - Melpomeni Fani
- Division of Radiopharmaceutical Chemistry, Department Theragnostics, University Hospital Basel, Basel 4031, Switzerland
| |
Collapse
|
15
|
Olari LR, Bauer R, Gil Miró M, Vogel V, Cortez Rayas L, Groß R, Gilg A, Klevesath R, Rodríguez Alfonso AA, Kaygisiz K, Rupp U, Pant P, Mieres-Pérez J, Steppe L, Schäffer R, Rauch-Wirth L, Conzelmann C, Müller JA, Zech F, Gerbl F, Bleher J, Preising N, Ständker L, Wiese S, Thal DR, Haupt C, Jonker HRA, Wagner M, Sanchez-Garcia E, Weil T, Stenger S, Fändrich M, von Einem J, Read C, Walther P, Kirchhoff F, Spellerberg B, Münch J. The C-terminal 32-mer fragment of hemoglobin alpha is an amyloidogenic peptide with antimicrobial properties. Cell Mol Life Sci 2023; 80:151. [PMID: 37198527 DOI: 10.1007/s00018-023-04795-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2022] [Accepted: 04/25/2023] [Indexed: 05/19/2023]
Abstract
Antimicrobial peptides (AMPs) are major components of the innate immune defense. Accumulating evidence suggests that the antibacterial activity of many AMPs is dependent on the formation of amyloid-like fibrils. To identify novel fibril forming AMPs, we generated a spleen-derived peptide library and screened it for the presence of amyloidogenic peptides. This approach led to the identification of a C-terminal 32-mer fragment of alpha-hemoglobin, termed HBA(111-142). The non-fibrillar peptide has membranolytic activity against various bacterial species, while the HBA(111-142) fibrils aggregated bacteria to promote their phagocytotic clearance. Further, HBA(111-142) fibrils selectively inhibited measles and herpes viruses (HSV-1, HSV-2, HCMV), but not SARS-CoV-2, ZIKV and IAV. HBA(111-142) is released from its precursor by ubiquitous aspartic proteases under acidic conditions characteristic at sites of infection and inflammation. Thus, HBA(111-142) is an amyloidogenic AMP that may specifically be generated from a highly abundant precursor during bacterial or viral infection and may play an important role in innate antimicrobial immune responses.
Collapse
Affiliation(s)
- Lia-Raluca Olari
- Institute of Molecular Virology, Ulm University Medical Center, 89081, Ulm, Germany
| | - Richard Bauer
- Institute of Medical Microbiology and Hygiene, Ulm University Medical Center, 89081, Ulm, Germany
| | - Marta Gil Miró
- Institute of Molecular Virology, Ulm University Medical Center, 89081, Ulm, Germany
| | - Verena Vogel
- Institute of Medical Microbiology and Hygiene, Ulm University Medical Center, 89081, Ulm, Germany
| | - Laura Cortez Rayas
- Institute of Virology, Ulm University Medical Center, 89081, Ulm, Germany
| | - Rüdiger Groß
- Institute of Molecular Virology, Ulm University Medical Center, 89081, Ulm, Germany
| | - Andrea Gilg
- Institute of Molecular Virology, Ulm University Medical Center, 89081, Ulm, Germany
| | - Raphael Klevesath
- Institute of Medical Microbiology and Hygiene, Ulm University Medical Center, 89081, Ulm, Germany
| | - Armando A Rodríguez Alfonso
- Core Facility for Functional Peptidomics, Ulm Peptide Pharmaceuticals (U-PEP), Ulm University Medical Center, 89081, Ulm, Germany
- Core Unit of Mass Spectrometry and Proteomics, Ulm University Medical Center, 89081, Ulm, Germany
| | - Kübra Kaygisiz
- Max-Planck-Institute for Polymer Research Mainz, 55128, Mainz, Germany
| | - Ulrich Rupp
- Central Facility for Electron Microscopy, Ulm University, 89081, Ulm, Germany
| | - Pradeep Pant
- Computational Biochemistry, Center of Medical Biotechnology, University of Duisburg-Essen, 45141, Essen, Germany
| | - Joel Mieres-Pérez
- Computational Biochemistry, Center of Medical Biotechnology, University of Duisburg-Essen, 45141, Essen, Germany
| | - Lena Steppe
- Institute of Molecular Virology, Ulm University Medical Center, 89081, Ulm, Germany
| | - Ramona Schäffer
- Institute of Molecular Virology, Ulm University Medical Center, 89081, Ulm, Germany
| | - Lena Rauch-Wirth
- Institute of Molecular Virology, Ulm University Medical Center, 89081, Ulm, Germany
| | - Carina Conzelmann
- Institute of Molecular Virology, Ulm University Medical Center, 89081, Ulm, Germany
| | - Janis A Müller
- Institute of Virology, Philipps University Marburg, 35043, Marburg, Germany
| | - Fabian Zech
- Institute of Molecular Virology, Ulm University Medical Center, 89081, Ulm, Germany
| | - Fabian Gerbl
- Institute of Medical Microbiology and Hygiene, Ulm University Medical Center, 89081, Ulm, Germany
| | - Jana Bleher
- Institute of Protein Biochemistry, Ulm University, 89081, Ulm, Germany
| | - Nico Preising
- Core Facility for Functional Peptidomics, Ulm Peptide Pharmaceuticals (U-PEP), Ulm University Medical Center, 89081, Ulm, Germany
| | - Ludger Ständker
- Core Facility for Functional Peptidomics, Ulm Peptide Pharmaceuticals (U-PEP), Ulm University Medical Center, 89081, Ulm, Germany
| | - Sebastian Wiese
- Core Unit of Mass Spectrometry and Proteomics, Ulm University Medical Center, 89081, Ulm, Germany
| | - Dietmar R Thal
- Laboratory of Neuropathology, Department of Imaging and Pathology, Leuven Brain Institute, KU Leuven, Leuven, Belgium
- Department of Pathology, UZ-Leuven, 3000, Leuven, Belgium
| | - Christian Haupt
- Institute of Protein Biochemistry, Ulm University, 89081, Ulm, Germany
| | - Hendrik R A Jonker
- Institute for Organic Chemistry and Chemical Biology, Center for Biomolecular Magnetic Resonance, Goethe University, 60438, Frankfurt am Main, Germany
| | - Manfred Wagner
- Max-Planck-Institute for Polymer Research Mainz, 55128, Mainz, Germany
| | - Elsa Sanchez-Garcia
- Computational Biochemistry, Center of Medical Biotechnology, University of Duisburg-Essen, 45141, Essen, Germany
| | - Tanja Weil
- Max-Planck-Institute for Polymer Research Mainz, 55128, Mainz, Germany
| | - Steffen Stenger
- Institute of Medical Microbiology and Hygiene, Ulm University Medical Center, 89081, Ulm, Germany
| | - Marcus Fändrich
- Institute of Protein Biochemistry, Ulm University, 89081, Ulm, Germany
| | - Jens von Einem
- Institute of Virology, Ulm University Medical Center, 89081, Ulm, Germany
| | - Clarissa Read
- Institute of Virology, Ulm University Medical Center, 89081, Ulm, Germany
- Central Facility for Electron Microscopy, Ulm University, 89081, Ulm, Germany
| | - Paul Walther
- Central Facility for Electron Microscopy, Ulm University, 89081, Ulm, Germany
| | - Frank Kirchhoff
- Institute of Molecular Virology, Ulm University Medical Center, 89081, Ulm, Germany
| | - Barbara Spellerberg
- Institute of Medical Microbiology and Hygiene, Ulm University Medical Center, 89081, Ulm, Germany
| | - Jan Münch
- Institute of Molecular Virology, Ulm University Medical Center, 89081, Ulm, Germany.
- Core Unit of Mass Spectrometry and Proteomics, Ulm University Medical Center, 89081, Ulm, Germany.
| |
Collapse
|
16
|
Gattringer J, Gruber CW, Hellinger R. Peptide modulators of cell migration: Overview, applications and future development. Drug Discov Today 2023; 28:103554. [PMID: 36921670 PMCID: PMC7615922 DOI: 10.1016/j.drudis.2023.103554] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2022] [Revised: 03/07/2023] [Accepted: 03/08/2023] [Indexed: 03/14/2023]
Abstract
Cell migration is a key physiological process in the development and homeostasis of multicellular organisms; errors in this complex system can trigger the development of cancer or inflammatory disorders. Therefore, modulating cell migration provides opportunities for drug discovery. Peptides are gaining importance on the global therapeutics market, given their unique properties compared with established small-molecule drugs or biologics. In this review, we identified over 470 peptides modulating cell migration and analyzed their characteristics. Over 95% of these peptides are in the discovery or preclinical stage, because the transition of peptide hits into drug leads often results in a bottleneck in the development process. We summarize chemical strategies in (pre-)clinical development to enhance drug-like properties of bioactive peptides.
Collapse
Affiliation(s)
- Jasmin Gattringer
- Medical University of Vienna, Center for Physiology and Pharmacology, Schwarzspanierstrasse 17, A-1090 Vienna, Austria
| | - Christian W Gruber
- Medical University of Vienna, Center for Physiology and Pharmacology, Schwarzspanierstrasse 17, A-1090 Vienna, Austria
| | - Roland Hellinger
- Medical University of Vienna, Center for Physiology and Pharmacology, Schwarzspanierstrasse 17, A-1090 Vienna, Austria.
| |
Collapse
|
17
|
Zhang M, Xu H. Peptide-assembled nanoparticles targeting tumor cells and tumor microenvironment for cancer therapy. Front Chem 2023; 11:1115495. [PMID: 36762192 PMCID: PMC9902599 DOI: 10.3389/fchem.2023.1115495] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2022] [Accepted: 01/09/2023] [Indexed: 01/26/2023] Open
Abstract
Tumor cells and corrupt stromal cells in the tumor microenvironment usually overexpress cancer-specific markers that are absent or barely detectable in normal cells, providing available targets for inhibiting the occurrence and development of cancers. It is noticeable that therapeutic peptides are emerging in cancer therapies and playing more and more important roles. Moreover, the peptides can be self-assembled and/or incorporated with polymeric molecules to form nanoparticles via non-covalent bond, which have presented appealing as well as enhanced capacities of recognizing targeted cells, responding to microenvironments, mediating internalization, and achieving therapeutic effects. In this review, we will introduce the peptide-based nanoparticles and their application advances in targeting tumor cells and stromal cells, including suppressive immune cells, fibrosis-related cells, and angiogenic vascular cells, for cancer therapy.
Collapse
|
18
|
Advanced EPI-X4 Derivatives Covalently Bind Human Serum Albumin Resulting in Prolonged Plasma Stability. Int J Mol Sci 2022; 23:ijms232315029. [PMID: 36499357 PMCID: PMC9735595 DOI: 10.3390/ijms232315029] [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: 10/04/2022] [Revised: 11/25/2022] [Accepted: 11/27/2022] [Indexed: 12/05/2022] Open
Abstract
Advanced derivatives of the Endogenous Peptide Inhibitor of CXCR4 (EPI-X4) have shown therapeutic efficacy upon topical administration in animal models of asthma and dermatitis. Here, we studied the plasma stability of the EPI-X4 lead compounds WSC02 and JM#21, using mass spectrometry to monitor the chemical integrity of the peptides and a functional fluorescence-based assay to determine peptide function in a CXCR4-antibody competition assay. Although mass spectrometry revealed very rapid disappearance of both peptides in human plasma within seconds, the functional assay revealed a significantly higher half-life of 9 min for EPI-X4 WSC02 and 6 min for EPI-X4 JM#21. Further analyses demonstrated that EPI-X4 WSC02 and EPI-X4 JM#21 interact with low molecular weight plasma components and serum albumin. Albumin binding is mediated by the formation of a disulfide bridge between Cys10 in the EPI-X4 peptides and Cys34 in albumin. These covalently linked albumin-peptide complexes have a higher stability in plasma as compared with the non-bound peptides and retain the ability to bind and antagonize CXCR4. Remarkably, chemically synthesized albumin-EPI-X4 conjugates coupled by non-breakable bonds have a drastically increased plasma stability of over 2 h. Thus, covalent coupling of EPI-X4 to albumin in vitro before administration or in vivo post administration may significantly increase the pharmacokinetic properties of this new class of CXCR4 antagonists.
Collapse
|
19
|
Monroe MK, Wang H, Anderson CF, Jia H, Flexner C, Cui H. Leveraging the therapeutic, biological, and self-assembling potential of peptides for the treatment of viral infections. J Control Release 2022; 348:1028-1049. [PMID: 35752254 PMCID: PMC11022941 DOI: 10.1016/j.jconrel.2022.06.037] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2022] [Revised: 06/06/2022] [Accepted: 06/20/2022] [Indexed: 12/12/2022]
Abstract
Peptides and peptide-based materials have an increasing role in the treatment of viral infections through their use as active pharmaceutical ingredients, targeting moieties, excipients, carriers, or structural components in drug delivery systems. The discovery of peptide-based therapeutic compounds, coupled with the development of new stabilization and formulation strategies, has led to a resurgence of antiviral peptide therapeutics over the past two decades. The ability of peptides to bind cell receptors and to facilitate membrane penetration and subsequent intracellular trafficking enables their use in various antiviral systems for improved targeting efficiency and treatment efficacy. Importantly, the self-assembly of peptides into well-defined nanostructures provides a vast library of discrete constructs and supramolecular biomaterials for systemic and local delivery of antiviral agents. We review here the recent progress in exploiting the therapeutic, biological, and self-assembling potential of peptides, peptide conjugates, and their supramolecular assemblies in treating human viral infections, with an emphasis on the treatment strategies for Human Immunodeficiency Virus (HIV).
Collapse
Affiliation(s)
- Maya K Monroe
- Department of Chemical and Biomolecular Engineering, The Johns Hopkins University, 3400 N Charles Street, Baltimore, MD 21218, United States of America; Institute for NanoBioTechnology, The Johns Hopkins University, 3400 N Charles Street, Baltimore, MD 21218, United States of America
| | - Han Wang
- Department of Chemical and Biomolecular Engineering, The Johns Hopkins University, 3400 N Charles Street, Baltimore, MD 21218, United States of America; Institute for NanoBioTechnology, The Johns Hopkins University, 3400 N Charles Street, Baltimore, MD 21218, United States of America
| | - Caleb F Anderson
- Department of Chemical and Biomolecular Engineering, The Johns Hopkins University, 3400 N Charles Street, Baltimore, MD 21218, United States of America; Institute for NanoBioTechnology, The Johns Hopkins University, 3400 N Charles Street, Baltimore, MD 21218, United States of America
| | - Hongpeng Jia
- Department of Surgery, The Johns Hopkins University School of Medicine, United States of America
| | - Charles Flexner
- Divisions of Clinical Pharmacology and Infectious Diseases, The Johns Hopkins University School of Medicine and Bloomberg School of Public Health, Baltimore, MD 21205, United States of America.
| | - Honggang Cui
- Department of Chemical and Biomolecular Engineering, The Johns Hopkins University, 3400 N Charles Street, Baltimore, MD 21218, United States of America; Institute for NanoBioTechnology, The Johns Hopkins University, 3400 N Charles Street, Baltimore, MD 21218, United States of America; Deptartment of Oncology and Sidney Kimmel Comprehensive Cancer Center, The Johns Hopkins University School of Medicine, Baltimore, MD 21205, United States of America; Center for Nanomedicine, The Wilmer Eye Institute, The Johns Hopkins University School of Medicine, 400 North Broadway, Baltimore, MD 21231, United States of America.
| |
Collapse
|
20
|
Romero-Molina S, Ruiz-Blanco YB, Mieres-Perez J, Harms M, Münch J, Ehrmann M, Sanchez-Garcia E. PPI-Affinity: A Web Tool for the Prediction and Optimization of Protein-Peptide and Protein-Protein Binding Affinity. J Proteome Res 2022; 21:1829-1841. [PMID: 35654412 PMCID: PMC9361347 DOI: 10.1021/acs.jproteome.2c00020] [Citation(s) in RCA: 38] [Impact Index Per Article: 12.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
Abstract
![]()
Virtual screening
of protein–protein and protein–peptide
interactions is a challenging task that directly impacts the processes
of hit identification and hit-to-lead optimization in drug design
projects involving peptide-based pharmaceuticals. Although several
screening tools designed to predict the binding affinity of protein–protein
complexes have been proposed, methods specifically developed to predict
protein–peptide binding affinity are comparatively scarce.
Frequently, predictors trained to score the affinity of small molecules
are used for peptides indistinctively, despite the larger complexity
and heterogeneity of interactions rendered by peptide binders. To
address this issue, we introduce PPI-Affinity, a tool that leverages
support vector machine (SVM) predictors of binding affinity to screen
datasets of protein–protein and protein–peptide complexes,
as well as to generate and rank mutants of a given structure. The
performance of the SVM models was assessed on four benchmark datasets,
which include protein–protein and protein–peptide binding
affinity data. In addition, we evaluated our model on a set of mutants
of EPI-X4, an endogenous peptide inhibitor of the chemokine receptor
CXCR4, and on complexes of the serine proteases HTRA1 and HTRA3 with
peptides. PPI-Affinity is freely accessible at https://protdcal.zmb.uni-due.de/PPIAffinity.
Collapse
Affiliation(s)
- Sandra Romero-Molina
- Computational Biochemistry, Center of Medical Biotechnology, University of Duisburg-Essen, Essen 45141, Germany
| | - Yasser B Ruiz-Blanco
- Computational Biochemistry, Center of Medical Biotechnology, University of Duisburg-Essen, Essen 45141, Germany
| | - Joel Mieres-Perez
- Computational Biochemistry, Center of Medical Biotechnology, University of Duisburg-Essen, Essen 45141, Germany
| | - Mirja Harms
- Institute of Molecular Virology, Ulm University Medical Center, Ulm 89081, Germany
| | - Jan Münch
- Institute of Molecular Virology, Ulm University Medical Center, Ulm 89081, Germany.,Core Facility Functional Peptidomics, Ulm University Medical Center, Ulm 89081, Germany
| | - Michael Ehrmann
- Faculty of Biology, Center of Medical Biotechnology, University of Duisburg-Essen, Essen 45141, Germany
| | - Elsa Sanchez-Garcia
- Computational Biochemistry, Center of Medical Biotechnology, University of Duisburg-Essen, Essen 45141, Germany
| |
Collapse
|
21
|
Caspar B, Cocchiara P, Melet A, Van Emelen K, Van der Aa A, Milligan G, Herbeuval JP. CXCR4 as a novel target in immunology: moving away from typical antagonists. FUTURE DRUG DISCOVERY 2022; 4:FDD77. [PMID: 35875591 PMCID: PMC9298491 DOI: 10.4155/fdd-2022-0007] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2022] [Accepted: 06/17/2022] [Indexed: 11/17/2022] Open
Abstract
CXCR4 has been a target of interest in drug discovery for numerous years. However, so far, most if not all studies focused on finding antagonists of CXCR4 function. Recent studies demonstrate that targeting a minor allosteric pocket of CXCR4 induces an immunomodulating effect in immune cells expressing CXCR4, connected to the TLR pathway. Compounds binding in this minor pocket seem to be functionally selective with inverse agonistic properties in selected GPCR signaling pathways (Gi activation), but additional signaling pathways are likely to be involved in the immunomodulating effects. In depth research into these CXCR4-targeted immunomodulators could lead to novel treatment options for (auto)-immune diseases.
Collapse
Affiliation(s)
- Birgit Caspar
- CNRS UMR-8601, 45 Rue des Saints-Pères, Paris, F-75006, France
- Team Chemistry & Biology, Modelling & Immunology for Therapy, CBMIT, Paris, France
- Université Paris Cité, CNRS, Laboratoire de Chimie et de Biochimie Pharmacologiques et Toxicologiques, Paris, F-75006, France
| | - Pietro Cocchiara
- Centre for Translational Pharmacology, Institute of Molecular, Cell & Systems Biology, College of Medical, Veterinary & Life Sciences, University of Glasgow, Glasgow, UK
| | - Armelle Melet
- CNRS UMR-8601, 45 Rue des Saints-Pères, Paris, F-75006, France
- Team Chemistry & Biology, Modelling & Immunology for Therapy, CBMIT, Paris, France
- Université Paris Cité, CNRS, Laboratoire de Chimie et de Biochimie Pharmacologiques et Toxicologiques, Paris, F-75006, France
| | - Kristof Van Emelen
- Ermium Therapeutics, Pépinière Paris Santé Cochin, 29 Rue du Faubourg Saint-Jacques, Paris, F-75014, France
| | - Annegret Van der Aa
- Ermium Therapeutics, Pépinière Paris Santé Cochin, 29 Rue du Faubourg Saint-Jacques, Paris, F-75014, France
| | - Graeme Milligan
- Centre for Translational Pharmacology, Institute of Molecular, Cell & Systems Biology, College of Medical, Veterinary & Life Sciences, University of Glasgow, Glasgow, UK
| | - Jean-Philippe Herbeuval
- CNRS UMR-8601, 45 Rue des Saints-Pères, Paris, F-75006, France
- Team Chemistry & Biology, Modelling & Immunology for Therapy, CBMIT, Paris, France
- Université Paris Cité, CNRS, Laboratoire de Chimie et de Biochimie Pharmacologiques et Toxicologiques, Paris, F-75006, France
| |
Collapse
|
22
|
Harms M, Hansson RF, Carmali S, Almeida-Hernández Y, Sanchez-Garcia E, Münch J, Zelikin AN. Dimerization of the Peptide CXCR4-Antagonist on Macromolecular and Supramolecular Protraction Arms Affords Increased Potency and Enhanced Plasma Stability. Bioconjug Chem 2022; 33:594-607. [PMID: 35293739 DOI: 10.1021/acs.bioconjchem.2c00034] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022]
Abstract
Peptides are prime drug candidates due to their high specificity of action but are disadvantaged by low proteolytic stability. Here, we focus on the development of stabilized analogues of EPI-X4, an endogenous peptide antagonist of CXCR4. We synthesized macromolecular peptide conjugates and performed side-by-side comparison with their albumin-binding counterparts and considered monovalent conjugates, divalent telechelic conjugates, and Y-shaped peptide dimers. All constructs were tested for competition with the CXCR4 antibody-receptor engagement, inhibition of receptor activation, and inhibition of the CXCR4-tropic human immunodeficiency virus infection. We found that the Y-shaped conjugates were more potent than the parent peptide and at the same time more stable in human plasma, with a favorable outlook for translational studies.
Collapse
Affiliation(s)
- Mirja Harms
- Institute of Molecular Virology, Ulm University Medical Center, 89081 Ulm, Germany
| | - Rikke Fabech Hansson
- Department of Chemistry and iNano Interdisciplinary Nanoscience Centre, Aarhus University, Aarhus 8000, Denmark
| | - Sheiliza Carmali
- Department of Chemistry and iNano Interdisciplinary Nanoscience Centre, Aarhus University, Aarhus 8000, Denmark
| | - Yasser Almeida-Hernández
- Computational Biochemistry, Center of Medical Biotechnology, University Duisburg-Essen, D-45141 Essen, Germany
| | - Elsa Sanchez-Garcia
- Computational Biochemistry, Center of Medical Biotechnology, University Duisburg-Essen, D-45141 Essen, Germany
| | - Jan Münch
- Institute of Molecular Virology, Ulm University Medical Center, 89081 Ulm, Germany
| | - Alexander N Zelikin
- Department of Chemistry and iNano Interdisciplinary Nanoscience Centre, Aarhus University, Aarhus 8000, Denmark
| |
Collapse
|
23
|
Endogenous Peptide Inhibitors of HIV Entry. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2022; 1366:65-85. [DOI: 10.1007/978-981-16-8702-0_5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/18/2022]
|
24
|
Schall N, Daubeuf F, Marsol C, Gizzi P, Frossard N, Bonnet D, Galzi JL, Muller S. A Selective Neutraligand for CXCL12/SDF-1α With Beneficial Regulatory Functions in MRL/Lpr Lupus Prone Mice. Front Pharmacol 2021; 12:752194. [PMID: 34744730 PMCID: PMC8566942 DOI: 10.3389/fphar.2021.752194] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2021] [Accepted: 09/28/2021] [Indexed: 11/13/2022] Open
Abstract
Dysregulation of CXCL12/SDF-1-CXCR4/CD184 signaling is associated with inflammatory diseases and notably with systemic lupus erythematosus. Issued from the lead molecule chalcone-4, the first neutraligand of the CXCL12 chemokine, LIT-927 was recently described as a potent analogue with improved solubility and stability. We aimed to investigate the capacity of LIT-927 to correct immune alterations in lupus-prone MRL/lpr mice and to explore the mechanism of action implemented by this small molecule in this model. We found that in contrast to AMD3100, an antagonist of CXCR4 and agonist of CXCR7, LIT-927 reduces the excessive number of several B/T lymphocyte subsets occurring in the blood of sick MRL/lpr mice (including CD3+/CD4-/CD8-/B220+ double negative T cells). In vitro, LIT-927 downregulated the overexpression of several activation markers on splenic MRL/lpr lymphocytes. It exerted effects on the CXCR4 pathway in MRL/lpr CD4+ T spleen cells. The results underline the importance of the CXCL12/CXCR4 axis in lupus pathophysiology. They indicate that neutralizing CXCL12 by the neutraligand LIT-927 can attenuate hyperactive lymphocytes in lupus. This mode of intervention might represent a novel strategy to control a common pathophysiological mechanism occurring in inflammatory diseases.
Collapse
Affiliation(s)
- Nicolas Schall
- CNRS UMR7242, Biotechnology and Cell Signaling, Ecole Supérieure de Biotechnologie de Strasbourg, Strasbourg University/Strasbourg Drug Discovery and Development Institute (IMS), Strasbourg, France
| | - François Daubeuf
- CNRS UMR7200, Laboratoire d'innovation Thérapeutique, Faculté de Pharmacie, Strasbourg University/Strasbourg Drug Discovery and Development Institute (IMS), Strasbourg, France.,CNRS UMS3286, Plate-forme de Chimie Biologique Intégrative de Strasbourg, Strasbourg University/ Strasbourg Drug Discovery and Development Institute (IMS), Strasbourg, France
| | - Claire Marsol
- CNRS UMR7200, Laboratoire d'innovation Thérapeutique, Faculté de Pharmacie, Strasbourg University/Strasbourg Drug Discovery and Development Institute (IMS), Strasbourg, France
| | - Patrick Gizzi
- CNRS UMS3286, Plate-forme de Chimie Biologique Intégrative de Strasbourg, Strasbourg University/ Strasbourg Drug Discovery and Development Institute (IMS), Strasbourg, France
| | - Nelly Frossard
- CNRS UMR7200, Laboratoire d'innovation Thérapeutique, Faculté de Pharmacie, Strasbourg University/Strasbourg Drug Discovery and Development Institute (IMS), Strasbourg, France
| | - Dominique Bonnet
- CNRS UMR7200, Laboratoire d'innovation Thérapeutique, Faculté de Pharmacie, Strasbourg University/Strasbourg Drug Discovery and Development Institute (IMS), Strasbourg, France
| | - Jean-Luc Galzi
- CNRS UMR7242, Biotechnology and Cell Signaling, Ecole Supérieure de Biotechnologie de Strasbourg, Strasbourg University/Strasbourg Drug Discovery and Development Institute (IMS), Strasbourg, France
| | - Sylviane Muller
- CNRS UMR7242, Biotechnology and Cell Signaling, Ecole Supérieure de Biotechnologie de Strasbourg, Strasbourg University/Strasbourg Drug Discovery and Development Institute (IMS), Strasbourg, France.,Fédération Hospitalo-Universitaire (FHU) OMICARE, Fédération de Médecine Translationnelle de Strasbourg (FMTS), Strasbourg University, Strasbourg, France.,University of Strasbourg Institute for Advanced Study (USIAS), Strasbourg, France
| |
Collapse
|
25
|
Pekmezovic M, Kaune AK, Austermeier S, Hitzler SUJ, Mogavero S, Hovhannisyan H, Gabaldón T, Gresnigt MS, Hube B. Human albumin enhances the pathogenic potential of Candida glabrata on vaginal epithelial cells. PLoS Pathog 2021; 17:e1010037. [PMID: 34710198 PMCID: PMC8577789 DOI: 10.1371/journal.ppat.1010037] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2021] [Revised: 11/09/2021] [Accepted: 10/15/2021] [Indexed: 12/15/2022] Open
Abstract
The opportunistic pathogen Candida glabrata is the second most frequent causative agent of vulvovaginal candidiasis (VVC), a disease that affects 70–75% of women at least once during their life. However, C. glabrata is almost avirulent in mice and normally incapable of inflicting damage to vaginal epithelial cells in vitro. We thus proposed that host factors present in vivo may influence C. glabrata pathogenicity. We, therefore, analyzed the impact of albumin, one of the most abundant proteins of the vaginal fluid. The presence of human, but not murine, albumin dramatically increased the potential of C. glabrata to damage vaginal epithelial cells. This effect depended on macropinocytosis-mediated epithelial uptake of albumin and subsequent proteolytic processing. The enhanced pathogenicity of C. glabrata can be explained by a combination of beneficial effects for the fungus, which includes an increased access to iron, accelerated growth, and increased adhesion. Screening of C. glabrata deletion mutants revealed that Hap5, a key regulator of iron homeostasis, is essential for the albumin-augmented damage potential. The albumin-augmented pathogenicity was reversed by the addition of iron chelators and a similar increase in pathogenicity was shown by increasing the iron availability, confirming a key role of iron. Accelerated growth not only led to higher cell numbers, but also to increased fungal metabolic activity and oxidative stress resistance. Finally, the albumin-driven enhanced damage potential was associated with the expression of distinct C. glabrata virulence genes. Transcriptional responses of the epithelial cells suggested an unfolded protein response (UPR) and ER-stress responses combined with glucose starvation induced by fast growing C. glabrata cells as potential mechanisms by which cytotoxicity is mediated.Collectively, we demonstrate that albumin augments the pathogenic potential of C. glabrata during interaction with vaginal epithelial cells. This suggests a role for albumin as a key player in the pathogenesis of VVC. Candida glabrata is the overall second causative species of candidiasis in humans, but little is known about the pathogenicity mechanisms of this yeast. C. glabrata is capable of causing lethal systemic candidiasis mostly in elderly immunocompromised patients, but is also a frequent cause of vulvovaginal candidiasis. These clinical insights suggest that C. glabrata has a high virulence potential, yet little pathogenicity is observed in both in vitro and in vivo infection models. The finding that human albumin, the most abundant protein in the human body, is boosting C. glabrata pathogenicity in vitro provides novel insights into C. glabrata pathogenicity mechanisms and shows that the presence of distinct human factors can have a significant influence on the virulence potential of a pathogenic microbe.
Collapse
Affiliation(s)
- Marina Pekmezovic
- Department of Microbial Pathogenicity Mechanisms, Leibniz Institute for Natural Product Research and Infection Biology, Hans Knoell Institute, Jena, Germany
| | - Ann-Kristin Kaune
- Department of Microbial Pathogenicity Mechanisms, Leibniz Institute for Natural Product Research and Infection Biology, Hans Knoell Institute, Jena, Germany
| | - Sophie Austermeier
- Department of Microbial Pathogenicity Mechanisms, Leibniz Institute for Natural Product Research and Infection Biology, Hans Knoell Institute, Jena, Germany
| | - Sophia U. J. Hitzler
- Junior Research Group Adaptive Pathogenicity Strategies, Leibniz Institute for Natural Product Research and Infection Biology, Hans Knoell Institute, Jena, Germany
| | - Selene Mogavero
- Department of Microbial Pathogenicity Mechanisms, Leibniz Institute for Natural Product Research and Infection Biology, Hans Knoell Institute, Jena, Germany
| | - Hrant Hovhannisyan
- Life Sciences Department, Barcelona Supercomputing Center (BSC), Barcelona, Spain
- Mechanisms of Disease Department, Institute for Research in Biomedicine (IRB), Barcelona, Spain
| | - Toni Gabaldón
- Life Sciences Department, Barcelona Supercomputing Center (BSC), Barcelona, Spain
- Mechanisms of Disease Department, Institute for Research in Biomedicine (IRB), Barcelona, Spain
- Institució Catalana de Recerca i Estudis Avançats (ICREA), Barcelona, Spain
| | - Mark S. Gresnigt
- Junior Research Group Adaptive Pathogenicity Strategies, Leibniz Institute for Natural Product Research and Infection Biology, Hans Knoell Institute, Jena, Germany
- * E-mail: (MSG); (BH)
| | - Bernhard Hube
- Department of Microbial Pathogenicity Mechanisms, Leibniz Institute for Natural Product Research and Infection Biology, Hans Knoell Institute, Jena, Germany
- Institute of Microbiology, Friedrich Schiller University, Jena, Germany
- * E-mail: (MSG); (BH)
| |
Collapse
|
26
|
Xu L, Silva MJSA, Gois PMP, Kuan SL, Weil T. Chemoselective cysteine or disulfide modification via single atom substitution in chloromethyl acryl reagents. Chem Sci 2021; 12:13321-13330. [PMID: 34777751 PMCID: PMC8528048 DOI: 10.1039/d1sc03250j] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2021] [Accepted: 09/06/2021] [Indexed: 12/19/2022] Open
Abstract
The development of bioconjugation chemistry has enabled the combination of various synthetic functionalities to proteins, giving rise to new classes of protein conjugates with functions well beyond what Nature can provide. Despite the progress in bioconjugation chemistry, there are no reagents developed to date where the reactivity can be tuned in a user-defined fashion to address different amino acid residues in proteins. Here, we report that 2-chloromethyl acryl reagents can serve as a simple yet versatile platform for selective protein modification at cysteine or disulfide sites by tuning their inherent electronic properties through the amide or ester linkage. Specifically, the 2-chloromethyl derivatives (acrylamide or acrylate) can be obtained via a simple and easily implemented one-pot reaction based on the coupling reaction between commercially available starting materials with different end-group functionalities (amino group or hydroxyl group). 2-Chloromethyl acrylamide reagents with an amide linkage favor selective modification at the cysteine site with fast reaction kinetics and near quantitative conversations. In contrast, 2-chloromethyl acrylate reagents bearing an ester linkage can undergo two successive Michael reactions, allowing the selective modification of disulfides bonds with high labeling efficiency and good conjugate stability. 2-Chloromethyl acryl derivatives (acrylamides and acrylates) can serve as simple and versatile bioconjugation reagents to achieve site-selective cysteine and disulfide modification on demand and with high efficiency.![]()
Collapse
Affiliation(s)
- Lujuan Xu
- Max Planck Institute for Polymer Research Ackermannweg 10 55128 Mainz Germany .,Institute of Inorganic Chemistry I, Ulm University Albert-Einstein-Allee 11 89081 Ulm Germany
| | - Maria J S A Silva
- Research Institute for Medicines (iMed.ULisboa), Faculty of Pharmacy, Universidade de Lisboa 1649-003 Lisbon Portugal
| | - Pedro M P Gois
- Research Institute for Medicines (iMed.ULisboa), Faculty of Pharmacy, Universidade de Lisboa 1649-003 Lisbon Portugal
| | - Seah Ling Kuan
- Max Planck Institute for Polymer Research Ackermannweg 10 55128 Mainz Germany .,Institute of Inorganic Chemistry I, Ulm University Albert-Einstein-Allee 11 89081 Ulm Germany
| | - Tanja Weil
- Max Planck Institute for Polymer Research Ackermannweg 10 55128 Mainz Germany .,Institute of Inorganic Chemistry I, Ulm University Albert-Einstein-Allee 11 89081 Ulm Germany
| |
Collapse
|
27
|
Rodríguez AA, Otero-González A, Ghattas M, Ständker L. Discovery, Optimization, and Clinical Application of Natural Antimicrobial Peptides. Biomedicines 2021; 9:1381. [PMID: 34680498 PMCID: PMC8533436 DOI: 10.3390/biomedicines9101381] [Citation(s) in RCA: 25] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2021] [Accepted: 09/28/2021] [Indexed: 12/12/2022] Open
Abstract
Antimicrobial peptides (AMPs) are widespread in multicellular organisms. These structurally diverse molecules are produced as the first line of defense against pathogens such as bacteria, viruses, fungi, and parasites. Also known as host defense peptides in higher eukaryotic organisms, AMPs display immunomodulatory and anticancer activities. During the last 30 years, technological advances have boosted the research on antimicrobial peptides, which have also attracted great interest as an alternative to tackling the antimicrobial resistance scenario mainly provoked by some bacterial and fungal pathogens. However, the introduction of natural AMPs in clinical trials faces challenges such as proteolytic digestion, short half-lives, and cytotoxicity upon systemic and oral application. Therefore, some strategies have been implemented to improve the properties of AMPs aiming to be used as effective therapeutic agents. In the present review, we summarize the discovery path of AMPs, focusing on preclinical development, recent advances in chemical optimization and peptide delivery systems, and their introduction into the market.
Collapse
Affiliation(s)
- Armando A. Rodríguez
- Core Facility for Functional Peptidomics, Ulm University Medical Center, 89081 Ulm, Germany
- Core Unit of Mass Spectrometry and Proteomics, Ulm University Medical Center, 89081 Ulm, Germany
| | | | - Maretchia Ghattas
- Faculty of Pharmacy and Biotechnology, German University in Cairo (GUC), Cairo 11511, Egypt;
| | - Ludger Ständker
- Core Facility for Functional Peptidomics, Ulm University Medical Center, 89081 Ulm, Germany
| |
Collapse
|
28
|
Sokkar P, Harms M, Stürzel C, Gilg A, Kizilsavas G, Raasholm M, Preising N, Wagner M, Kirchhoff F, Ständker L, Weidinger G, Mayer B, Münch J, Sanchez-Garcia E. Computational modeling and experimental validation of the EPI-X4/CXCR4 complex allows rational design of small peptide antagonists. Commun Biol 2021; 4:1113. [PMID: 34552197 PMCID: PMC8458281 DOI: 10.1038/s42003-021-02638-5] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2021] [Accepted: 08/31/2021] [Indexed: 11/13/2022] Open
Abstract
EPI-X4, a 16-mer fragment of albumin, is a specific endogenous antagonist and inverse agonist of the CXC-motif-chemokine receptor 4 (CXCR4) and thus a key regulator of CXCR4 function. Accordingly, activity-optimized synthetic derivatives of EPI-X4 are promising leads for the therapy of CXCR4-linked disorders such as cancer or inflammatory diseases. We investigated the binding of EPI-X4 to CXCR4, which so far remained unclear, by means of biomolecular simulations combined with experimental mutagenesis and activity studies. We found that EPI-X4 interacts through its N-terminal residues with CXCR4 and identified its key interaction motifs, explaining receptor antagonization. Using this model, we developed shortened EPI-X4 derivatives (7-mers) with optimized receptor antagonizing properties as new leads for the development of CXCR4 inhibitors. Our work reveals the molecular details and mechanism by which the first endogenous peptide antagonist of CXCR4 interacts with its receptor and provides a foundation for the rational design of improved EPI-X4 derivatives.
Collapse
Affiliation(s)
- Pandian Sokkar
- Computational Biochemistry, Center of Medical Biotechnology, University of Duisburg-Essen, Essen, Germany
- Faculty of Allied Health Sciences, Chettinad Hospital and Research Institute, Chettinad Academy of Research and Education, Kelambakkam, Tamil Nadu, 603103, India
| | - Mirja Harms
- Institute of Molecular Virology, Ulm University Medical Center, Ulm, Germany
| | - Christina Stürzel
- Institute of Molecular Virology, Ulm University Medical Center, Ulm, Germany
| | - Andrea Gilg
- Institute of Molecular Virology, Ulm University Medical Center, Ulm, Germany
| | | | - Martina Raasholm
- Institute of Biochemistry and Molecular Biology, Ulm University, Ulm, 89081, Germany
| | - Nico Preising
- Core Facility Functional Peptidomics, Ulm University Medical Center, Ulm, 89081, Germany
| | - Manfred Wagner
- Max Planck Institute for Polymer Research, Mainz, Germany
| | - Frank Kirchhoff
- Institute of Molecular Virology, Ulm University Medical Center, Ulm, Germany
| | - Ludger Ständker
- Core Facility Functional Peptidomics, Ulm University Medical Center, Ulm, 89081, Germany
| | - Gilbert Weidinger
- Institute of Biochemistry and Molecular Biology, Ulm University, Ulm, 89081, Germany
| | - Benjamin Mayer
- Institute for Epidemiology and Medical Biometry, Ulm University, Ulm, 89075, Germany
| | - Jan Münch
- Institute of Molecular Virology, Ulm University Medical Center, Ulm, Germany.
- Core Facility Functional Peptidomics, Ulm University Medical Center, Ulm, 89081, Germany.
| | - Elsa Sanchez-Garcia
- Computational Biochemistry, Center of Medical Biotechnology, University of Duisburg-Essen, Essen, Germany.
| |
Collapse
|
29
|
Harms M, Habib MM, Nemska S, Nicolò A, Gilg A, Preising N, Sokkar P, Carmignani S, Raasholm M, Weidinger G, Kizilsavas G, Wagner M, Ständker L, Abadi AH, Jumaa H, Kirchhoff F, Frossard N, Sanchez-Garcia E, Münch J. An optimized derivative of an endogenous CXCR4 antagonist prevents atopic dermatitis and airway inflammation. Acta Pharm Sin B 2021; 11:2694-2708. [PMID: 34589390 PMCID: PMC8463264 DOI: 10.1016/j.apsb.2020.12.005] [Citation(s) in RCA: 31] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2020] [Revised: 11/10/2020] [Accepted: 12/01/2020] [Indexed: 02/07/2023] Open
Abstract
Aberrant CXCR4/CXCL12 signaling is involved in many pathophysiological processes such as cancer and inflammatory diseases. A natural fragment of serum albumin, named EPI-X4, has previously been identified as endogenous peptide antagonist and inverse agonist of CXCR4 and is a promising compound for the development of improved analogues for the therapy of CXCR4-associated diseases. To generate optimized EPI-X4 derivatives we here performed molecular docking analysis to identify key interaction motifs of EPI-X4/CXCR4. Subsequent rational drug design allowed to increase the anti-CXCR4 activity of EPI-X4. The EPI-X4 derivative JM#21 bound CXCR4 and suppressed CXCR4-tropic HIV-1 infection more efficiently than the clinically approved small molecule CXCR4 antagonist AMD3100. EPI-X4 JM#21 did not exert toxic effects in zebrafish embryos and suppressed allergen-induced infiltration of eosinophils and other immune cells into the airways of animals in an asthma mouse model. Moreover, topical administration of the optimized EPI-X4 derivative efficiently prevented inflammation of the skin in a mouse model of atopic dermatitis. Thus, rationally designed EPI-X4 JM#21 is a novel potent antagonist of CXCR4 and the first CXCR4 inhibitor with therapeutic efficacy in atopic dermatitis. Further clinical development of this new class of CXCR4 antagonists for the therapy of atopic dermatitis, asthma and other CXCR4-associated diseases is highly warranted.
Collapse
|
30
|
Neidleman J, Luo X, George AF, McGregor M, Yang J, Yun C, Murray V, Gill G, Greene WC, Vasquez J, Lee SA, Ghosn E, Lynch KL, Roan NR. Distinctive features of SARS-CoV-2-specific T cells predict recovery from severe COVID-19. Cell Rep 2021; 36:109414. [PMID: 34260965 PMCID: PMC8238659 DOI: 10.1016/j.celrep.2021.109414] [Citation(s) in RCA: 63] [Impact Index Per Article: 15.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2021] [Revised: 05/14/2021] [Accepted: 06/24/2021] [Indexed: 02/08/2023] Open
Abstract
Although T cells are likely players in severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) immunity, little is known about the phenotypic features of SARS-CoV-2-specific T cells associated with recovery from severe coronavirus disease 2019 (COVID-19). We analyze T cells from 34 individuals with COVID-19 with severity ranging from mild (outpatient) to critical, culminating in death. Relative to individuals who succumbed, individuals who recovered from severe COVID-19 harbor elevated and increasing numbers of SARS-CoV-2-specific T cells capable of homeostatic proliferation. In contrast, fatal COVID-19 cases display elevated numbers of SARS-CoV-2-specific regulatory T cells and a time-dependent escalation in activated bystander CXCR4+ T cells, as assessed by longitudinal sampling. Together with the demonstration of increased proportions of inflammatory CXCR4+ T cells in the lungs of individuals with severe COVID-19, these results support a model where lung-homing T cells activated through bystander effects contribute to immunopathology, whereas a robust, non-suppressive SARS-CoV-2-specific T cell response limits pathogenesis and promotes recovery from severe COVID-19.
Collapse
Affiliation(s)
- Jason Neidleman
- Gladstone Institutes, San Francisco, CA 94158, USA; Department of Urology, University of California, San Francisco, CA 94158, USA
| | - Xiaoyu Luo
- Gladstone Institutes, San Francisco, CA 94158, USA
| | - Ashley F George
- Gladstone Institutes, San Francisco, CA 94158, USA; Department of Urology, University of California, San Francisco, CA 94158, USA
| | - Matthew McGregor
- Gladstone Institutes, San Francisco, CA 94158, USA; Department of Urology, University of California, San Francisco, CA 94158, USA
| | - Junkai Yang
- Deptartments of Medicine and Pediatrics, Lowance Center for Human Immunology, Emory Vaccine Center, Emory University, Atlanta, GA 30322, USA
| | - Cassandra Yun
- Department of Laboratory Medicine, University of California, San Francisco, CA 94110, USA
| | - Victoria Murray
- Zuckerberg San Francisco General Hospital and the University of California, San Francisco, CA 94110, USA
| | - Gurjot Gill
- Zuckerberg San Francisco General Hospital and the University of California, San Francisco, CA 94110, USA
| | - Warner C Greene
- Gladstone Institutes, San Francisco, CA 94158, USA; Department of Medicine, University of California, San Francisco, CA 94110, USA
| | - Joshua Vasquez
- Department of Medicine, University of California, San Francisco, CA 94110, USA
| | - Sulggi A Lee
- Zuckerberg San Francisco General Hospital and the University of California, San Francisco, CA 94110, USA
| | - Eliver Ghosn
- Deptartments of Medicine and Pediatrics, Lowance Center for Human Immunology, Emory Vaccine Center, Emory University, Atlanta, GA 30322, USA.
| | - Kara L Lynch
- Department of Laboratory Medicine, University of California, San Francisco, CA 94110, USA.
| | - Nadia R Roan
- Gladstone Institutes, San Francisco, CA 94158, USA; Department of Urology, University of California, San Francisco, CA 94158, USA.
| |
Collapse
|
31
|
Holch A, Bauer R, Olari LR, Rodriguez AA, Ständker L, Preising N, Karacan M, Wiese S, Walther P, Ruiz-Blanco YB, Sanchez-Garcia E, Schumann C, Münch J, Spellerberg B. Respiratory ß-2-Microglobulin exerts pH dependent antimicrobial activity. Virulence 2021; 11:1402-1414. [PMID: 33092477 PMCID: PMC7588194 DOI: 10.1080/21505594.2020.1831367] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
The respiratory tract is a major entry site for microbial pathogens. To combat bacterial infections, the immune system has various defense mechanisms at its disposal, including antimicrobial peptides (AMPs). To search for novel AMPs from the respiratory tract, a peptide library from human broncho-alveolar-lavage (BAL) fluid was screened for antimicrobial activity by radial diffusion assays allowing the efficient detection of antibacterial activity within a small sample size. After repeated testing-cycles and subsequent purification, we identified ß-2-microglobulin (B2M) in antibacterially active fractions. B2M belongs to the MHC-1 receptor complex present at the surface of nucleated cells. It is known to inhibit the growth of Listeria monocytogenes and Escherichia coli and to facilitate phagocytosis of Staphylococcus aureus. Using commercially available B2M we confirmed a dose-dependent inhibition of Pseudomonas aeruginosa and L. monocytogenes. To characterize AMP activity within the B2M sequence, peptide fragments of the molecule were tested for antimicrobial activity. Activity could be localized to the C-terminal part of B2M. Investigating pH dependency of the antimicrobial activity of B2M demonstrated an increased activity at pH values of 5.5 and below, a hallmark of infection and inflammation. Sytox green uptake into bacterial cells following the exposure to B2M was determined and revealed a pH-dependent loss of bacterial membrane integrity. TEM analysis showed areas of disrupted bacterial membranes in L. monocytogenes incubated with B2M and high amounts of lysed bacterial cells. In conclusion, B2M as part of a ubiquitous cell surface complex may represent a potent antimicrobial agent by interfering with bacterial membrane integrity.
Collapse
Affiliation(s)
- Armin Holch
- Institute of Medical Microbiology and Hygiene, University Hospital , Ulm, Germany
| | - Richard Bauer
- Institute of Medical Microbiology and Hygiene, University Hospital , Ulm, Germany
| | - Lia-Raluca Olari
- Institute of Molecular Virology, University Hospital , Ulm, Germany
| | - Armando A Rodriguez
- Core Facility Functional Peptidomics, Ulm University Medical Center , Ulm, Germany.,Core Unit Mass Spectrometry and Proteomics, Ulm University , Ulm, Germany
| | - Ludger Ständker
- Core Facility Functional Peptidomics, Ulm University Medical Center , Ulm, Germany
| | - Nico Preising
- Core Facility Functional Peptidomics, Ulm University Medical Center , Ulm, Germany
| | - Merve Karacan
- Core Facility Functional Peptidomics, Ulm University Medical Center , Ulm, Germany
| | - Sebastian Wiese
- Core Unit Mass Spectrometry and Proteomics, Ulm University , Ulm, Germany
| | - Paul Walther
- Central Facility for Electron Microscopy, Ulm University Medical Center , Ulm, Germany
| | - Yasser B Ruiz-Blanco
- Computational Biochemistry, Center of Medical Biotechnology, University of Duisburg-Essen , Essen, Germany
| | - Elsa Sanchez-Garcia
- Computational Biochemistry, Center of Medical Biotechnology, University of Duisburg-Essen , Essen, Germany
| | - Christian Schumann
- Pneumology, Thoracic Oncology, Sleep and Respiratory Critical Care Medicine, Clinics Kempten-Allgäu, Kempten and Immenstadt , Germany
| | - Jan Münch
- Institute of Molecular Virology, University Hospital , Ulm, Germany.,Core Facility Functional Peptidomics, Ulm University Medical Center , Ulm, Germany
| | - Barbara Spellerberg
- Institute of Medical Microbiology and Hygiene, University Hospital , Ulm, Germany
| |
Collapse
|
32
|
Beitzinger B, Gerbl F, Vomhof T, Schmid R, Noschka R, Rodriguez A, Wiese S, Weidinger G, Ständker L, Walther P, Michaelis J, Lindén M, Stenger S. Delivery by Dendritic Mesoporous Silica Nanoparticles Enhances the Antimicrobial Activity of a Napsin-Derived Peptide Against Intracellular Mycobacterium tuberculosis. Adv Healthc Mater 2021; 10:e2100453. [PMID: 34142469 PMCID: PMC11468746 DOI: 10.1002/adhm.202100453] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2021] [Revised: 05/20/2021] [Indexed: 12/28/2022]
Abstract
Tuberculosis remains a serious global health problem causing 1.3 million deaths annually. The causative pathogen Mycobacterium tuberculosis (Mtb) has developed several mechanisms to evade the immune system and resistances to many conventional antibiotics, so that alternative treatment strategies are urgently needed. By isolation from bronchoalveolar lavage and peptide optimization, a new antimicrobial peptide named NapFab is discovered. While showing robust activity against extracellular Mtb, the activity of NapFab against intracellular bacteria is limited due to low intracellular availability. By loading NapFab onto dendritic mesoporous silica nanoparticles (DMSN) as a carrier system, cellular uptake, and consequently antimycobacterial activity against intracellular Mtb is significantly enhanced. Furthermore, using lattice light-sheet fluorescence microscopy, it can be shown that the peptide is gradually released from the DMSN inside living macrophages over time. By electron microscopy and tomography, it is demonstrated that peptide loaded DMSN are stored in vesicular structures in proximity to mycobacterial phagosomes inside the cells, but the nanoparticles are typically not in direct contact with the bacteria. Based on the combination of functional and live-cell imaging analyses, it is hypothesized that after being released from the DMSN NapFab is able to enter the bacterial phagosome and gain access to the bacilli.
Collapse
Affiliation(s)
- Bastian Beitzinger
- Institute of Inorganic Chemistry IIUlm UniversityAlbert‐Einstein‐Allee 11Ulm89081Germany
| | - Fabian Gerbl
- Institute of Medical Microbiology and HygieneUlm University HospitalAlbert‐Einstein‐Allee 11Ulm89081Germany
| | - Thomas Vomhof
- Institute of BiophysicsUlm UniversityAlbert‐Einstein‐Allee 11Ulm89081Germany
| | - Roman Schmid
- Institute of Inorganic Chemistry IIUlm UniversityAlbert‐Einstein‐Allee 11Ulm89081Germany
| | - Reiner Noschka
- Institute of Medical Microbiology and HygieneUlm University HospitalAlbert‐Einstein‐Allee 11Ulm89081Germany
| | - Armando Rodriguez
- Core Facility of Functional PeptidomicsUlm UniversityMeyerhofstraße 4Ulm89081Germany
- Core Unit of Mass Spectrometry and ProteomicsUlm UniversityAlbert‐Einstein Allee 11Ulm89081Germany
| | - Sebastian Wiese
- Core Unit of Mass Spectrometry and ProteomicsUlm UniversityAlbert‐Einstein Allee 11Ulm89081Germany
| | - Gilbert Weidinger
- Institute of Biochemistry and Molecular BiologyUlm UniversityAlbert‐Einstein‐Allee 11Ulm89081Germany
| | - Ludger Ständker
- Core Facility of Functional PeptidomicsUlm UniversityMeyerhofstraße 4Ulm89081Germany
| | - Paul Walther
- Central Facility for Electron MicroscopyUlm UniversityAlbert‐Einstein‐Allee 11Ulm89081Germany
| | - Jens Michaelis
- Institute of BiophysicsUlm UniversityAlbert‐Einstein‐Allee 11Ulm89081Germany
| | - Mika Lindén
- Institute of Inorganic Chemistry IIUlm UniversityAlbert‐Einstein‐Allee 11Ulm89081Germany
| | - Steffen Stenger
- Institute of Medical Microbiology and HygieneUlm University HospitalAlbert‐Einstein‐Allee 11Ulm89081Germany
| |
Collapse
|
33
|
Cano-Garrido O, Álamo P, Sánchez-García L, Falgàs A, Sánchez-Chardi A, Serna N, Parladé E, Unzueta U, Roldán M, Voltà-Durán E, Casanova I, Villaverde A, Mangues R, Vázquez E. Biparatopic Protein Nanoparticles for the Precision Therapy of CXCR4 + Cancers. Cancers (Basel) 2021; 13:2929. [PMID: 34208189 PMCID: PMC8230831 DOI: 10.3390/cancers13122929] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2021] [Revised: 05/28/2021] [Accepted: 06/07/2021] [Indexed: 01/05/2023] Open
Abstract
The accumulated molecular knowledge about human cancer enables the identification of multiple cell surface markers as highly specific therapeutic targets. A proper tumor targeting could significantly avoid drug exposure of healthy cells, minimizing side effects, but it is also expected to increase the therapeutic index. Specifically, colorectal cancer has a particularly poor prognosis in late stages, being drug targeting an appropriate strategy to substantially improve the therapeutic efficacy. In this study, we have explored the potential of the human albumin-derived peptide, EPI-X4, as a suitable ligand to target colorectal cancer via the cell surface protein CXCR4, a chemokine receptor overexpressed in cancer stem cells. To explore the potential use of this ligand, self-assembling protein nanoparticles have been generated displaying an engineered EPI-X4 version, which conferred a modest CXCR4 targeting and fast and high level of cell apoptosis in tumor CXCR4+ cells, in vitro and in vivo. In addition, when EPI-X4-based building blocks are combined with biologically inert polypeptides containing the CXCR4 ligand T22, the resulting biparatopic nanoparticles show a dramatically improved biodistribution in mouse models of CXCR4+ human cancer, faster cell internalization and enhanced target cell death when compared to the version based on a single ligand. The generation of biparatopic materials opens exciting possibilities in oncotherapies based on high precision drug delivery based on the receptor CXCR4.
Collapse
Affiliation(s)
- Olivia Cano-Garrido
- Nanoligent SL, Edifici EUREKA, Universitat Autònoma de Barcelona, Bellaterra, 08193 Barcelona, Spain
- Institut de Biotecnologia i de Biomedicina, Universitat Autònoma de Barcelona, Bellaterra, 08193 Barcelona, Spain
| | - Patricia Álamo
- CIBER de Bioingeniería, Biomateriales y Nanomedicina (CIBER-BBN), C/Monforte de Lemos 3-5, 28029 Madrid, Spain
- Instituto de Investigación Biomédica Sant Pau (IIB Sant Pau), Sant Antoni Ma Claret 167, 08025 Barcelona, Spain
- Instituto de Investigación Contra la Leucemia Josep Carreras, 08025 Barcelona, Spain
| | - Laura Sánchez-García
- Institut de Biotecnologia i de Biomedicina, Universitat Autònoma de Barcelona, Bellaterra, 08193 Barcelona, Spain
- CIBER de Bioingeniería, Biomateriales y Nanomedicina (CIBER-BBN), C/Monforte de Lemos 3-5, 28029 Madrid, Spain
- Departament de Genètica i de Microbiologia, Universitat Autònoma de Barcelona, Bellaterra, 08193 Barcelona, Spain
| | - Aïda Falgàs
- CIBER de Bioingeniería, Biomateriales y Nanomedicina (CIBER-BBN), C/Monforte de Lemos 3-5, 28029 Madrid, Spain
- Instituto de Investigación Biomédica Sant Pau (IIB Sant Pau), Sant Antoni Ma Claret 167, 08025 Barcelona, Spain
- Instituto de Investigación Contra la Leucemia Josep Carreras, 08025 Barcelona, Spain
| | - Alejandro Sánchez-Chardi
- Departament de Biologia Evolutiva, Ecologia i Ciències Ambientals, Facultat de Biologia, Universitat de Barcelona, Av. Diagonal 643, 08028 Barcelona, Spain
- Servei de Microscòpia, Universitat Autònoma de Barcelona, Bellaterra, 08193 Barcelona, Spain
| | - Naroa Serna
- Nanoligent SL, Edifici EUREKA, Universitat Autònoma de Barcelona, Bellaterra, 08193 Barcelona, Spain
- Institut de Biotecnologia i de Biomedicina, Universitat Autònoma de Barcelona, Bellaterra, 08193 Barcelona, Spain
- CIBER de Bioingeniería, Biomateriales y Nanomedicina (CIBER-BBN), C/Monforte de Lemos 3-5, 28029 Madrid, Spain
| | - Eloi Parladé
- Institut de Biotecnologia i de Biomedicina, Universitat Autònoma de Barcelona, Bellaterra, 08193 Barcelona, Spain
- CIBER de Bioingeniería, Biomateriales y Nanomedicina (CIBER-BBN), C/Monforte de Lemos 3-5, 28029 Madrid, Spain
| | - Ugutz Unzueta
- CIBER de Bioingeniería, Biomateriales y Nanomedicina (CIBER-BBN), C/Monforte de Lemos 3-5, 28029 Madrid, Spain
- Instituto de Investigación Biomédica Sant Pau (IIB Sant Pau), Sant Antoni Ma Claret 167, 08025 Barcelona, Spain
- Instituto de Investigación Contra la Leucemia Josep Carreras, 08025 Barcelona, Spain
- Departament de Genètica i de Microbiologia, Universitat Autònoma de Barcelona, Bellaterra, 08193 Barcelona, Spain
| | - Mònica Roldán
- Unitat de Microscòpia Confocal i Imatge Cel·lular, Servei de Medicina Genètica i Molecular, Institut Pediàtric de Malalties Rares (IPER), Hospital Sant Joan de Déu, Esplugues de Llobregat, 08950 Barcelona, Spain
- Institut de Recerca Sant Joan de Déu, Esplugues de Llobregat, 08950 Barcelona, Spain
| | - Eric Voltà-Durán
- Institut de Biotecnologia i de Biomedicina, Universitat Autònoma de Barcelona, Bellaterra, 08193 Barcelona, Spain
- CIBER de Bioingeniería, Biomateriales y Nanomedicina (CIBER-BBN), C/Monforte de Lemos 3-5, 28029 Madrid, Spain
- Departament de Genètica i de Microbiologia, Universitat Autònoma de Barcelona, Bellaterra, 08193 Barcelona, Spain
| | - Isolda Casanova
- CIBER de Bioingeniería, Biomateriales y Nanomedicina (CIBER-BBN), C/Monforte de Lemos 3-5, 28029 Madrid, Spain
- Instituto de Investigación Biomédica Sant Pau (IIB Sant Pau), Sant Antoni Ma Claret 167, 08025 Barcelona, Spain
- Instituto de Investigación Contra la Leucemia Josep Carreras, 08025 Barcelona, Spain
| | - Antonio Villaverde
- Institut de Biotecnologia i de Biomedicina, Universitat Autònoma de Barcelona, Bellaterra, 08193 Barcelona, Spain
- CIBER de Bioingeniería, Biomateriales y Nanomedicina (CIBER-BBN), C/Monforte de Lemos 3-5, 28029 Madrid, Spain
- Departament de Genètica i de Microbiologia, Universitat Autònoma de Barcelona, Bellaterra, 08193 Barcelona, Spain
| | - Ramón Mangues
- CIBER de Bioingeniería, Biomateriales y Nanomedicina (CIBER-BBN), C/Monforte de Lemos 3-5, 28029 Madrid, Spain
- Instituto de Investigación Biomédica Sant Pau (IIB Sant Pau), Sant Antoni Ma Claret 167, 08025 Barcelona, Spain
- Instituto de Investigación Contra la Leucemia Josep Carreras, 08025 Barcelona, Spain
| | - Esther Vázquez
- Institut de Biotecnologia i de Biomedicina, Universitat Autònoma de Barcelona, Bellaterra, 08193 Barcelona, Spain
- CIBER de Bioingeniería, Biomateriales y Nanomedicina (CIBER-BBN), C/Monforte de Lemos 3-5, 28029 Madrid, Spain
- Departament de Genètica i de Microbiologia, Universitat Autònoma de Barcelona, Bellaterra, 08193 Barcelona, Spain
| |
Collapse
|
34
|
Gilg A, Harms M, Olari LR, Urbanowitz AK, Bonig H, Münch J. Absence of the CXCR4 antagonist EPI-X4 from pharmaceutical human serum albumin preparations. J Transl Med 2021; 19:190. [PMID: 33941197 PMCID: PMC8094565 DOI: 10.1186/s12967-021-02859-6] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2021] [Accepted: 04/26/2021] [Indexed: 11/18/2022] Open
Abstract
BACKGROUND Endogenous Peptide Inhibitor of CXCR4 (EPI-X4) is a natural antagonist of the CXC chemokine receptor 4 (CXCR4). EPI-X4 is a 16-mer peptide that is released from human serum albumin (HSA) by acidic aspartic proteases such as Cathepsin D and E. Since human serum albumin (HSA) is an important medicinal substance we asked whether different pharmaceutical HSA products contain EPI-X4 which could have been generated during manufacturing and whether HSA can serve as a substrate for cathepsins despite of the presence of stabilizers like caprylate. METHODS Eight pharmaceutical HSA preparations representing all currently used fractionation technologies were analyzed. The previously described specific EPI-X4 ELISA was used for quantification; in vitro EPI-X4 generation by acidification in the presence or absence of cathepsins was followed by quantification with ELISA. RESULTS None of the pharmaceutical HSA preparations tested contained EPI-X4. Acidification of HSA did not generate EPI-X4. Addition of cathepsins D and E to acidified HSA yielded high concentrations of EPI-X4 in all HSA preparations, indistinguishable between individual products. CONCLUSION Medicinal HSA preparations per se do not contain EPI-X4, but will replenish its precursor which can be cleaved to EPI-X4 in vivo, environmental conditions permitting.
Collapse
Affiliation(s)
- Andrea Gilg
- Institute of Molecular Virology, Ulm University Medical Center, 89081, Ulm, Germany
| | - Mirja Harms
- Institute of Molecular Virology, Ulm University Medical Center, 89081, Ulm, Germany
| | - Lia-Raluca Olari
- Institute of Molecular Virology, Ulm University Medical Center, 89081, Ulm, Germany
| | - Ann-Kathrin Urbanowitz
- German Red Cross Blood Donor Service Baden-Wuerttemberg-Hessen, Institute Frankfurt, 60528, Frankfurt, Germany
| | - Halvard Bonig
- German Red Cross Blood Donor Service Baden-Wuerttemberg-Hessen, Institute Frankfurt, 60528, Frankfurt, Germany.
- Institute for Transfusion Medicine and Immunohematology, Goethe University, 60528, Frankfurt, Germany.
- Dept. of Medicine, Div. of Hematology, University of Washington, Seattle, WA, 98195, USA.
| | - Jan Münch
- Institute of Molecular Virology, Ulm University Medical Center, 89081, Ulm, Germany.
- Core Facility Functional Peptidomics, Ulm University Medical Center, 89081, Ulm, Germany.
| |
Collapse
|
35
|
Kaiser LM, Harms M, Sauter D, Rawat VPS, Glitscher M, Hildt E, Tews D, Hunter Z, Münch J, Buske C. Targeting of CXCR4 by the Naturally Occurring CXCR4 Antagonist EPI-X4 in Waldenström's Macroglobulinemia. Cancers (Basel) 2021; 13:826. [PMID: 33669329 PMCID: PMC7920274 DOI: 10.3390/cancers13040826] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2020] [Revised: 02/05/2021] [Accepted: 02/11/2021] [Indexed: 12/31/2022] Open
Abstract
CXCR4 expression and downstream signaling have been identified as key factors in malignant hematopoiesis. Thus, up to 40% of all patients with Waldenström's macroglobulinemia (WM) carry an activating mutation of CXCR4 that leads to a more aggressive clinical course and inferior outcome upon treatment with the Bruton's tyrosine kinase inhibitor ibrutinib. Nevertheless, little is known about physiological mechanisms counteracting CXCR4 signaling in hematopoietic neoplasms. Recently, the endogenous human peptide EPI-X4 was identified as a natural CXCR4 antagonist that effectively blocks CXCL12-mediated receptor internalization and suppresses the migration and invasion of cancer cells towards a CXCL12 gradient. Here, we demonstrate that EPI-X4 efficiently binds to CXCR4 of WM cells and decreases their migration towards CXCL12. The CXCR4 inhibitory activity of EPI-X4 is accompanied by reduced expression of genes involved in MAPK signaling and energy metabolism. Notably, the anti-WM activity of EPI-X4 could be further augmented by the rational design of EPI-X4 derivatives showing higher binding affinity to CXCR4. In summary, these data demonstrate that a naturally occurring anti-CXCR4 peptide is able to interfere with WM cell behaviour, and that optimized derivatives of EPI-X4 may represent a promising approach in suppressing growth promoting CXCR4 signaling in WM.
Collapse
Affiliation(s)
- Lisa Marie Kaiser
- Comprehensive Cancer Center Ulm, Institute of Experimental Cancer Research, University Hospital Ulm, 89081 Ulm, Germany; (L.M.K.); (V.P.S.R.)
| | - Mirja Harms
- Institute of Molecular Virology, Ulm University Medical Center, 89081 Ulm, Germany; (M.H.); (D.S.); (J.M.)
| | - Daniel Sauter
- Institute of Molecular Virology, Ulm University Medical Center, 89081 Ulm, Germany; (M.H.); (D.S.); (J.M.)
- Institute for Medical Virology and Epidemiology of Viral Diseases, University Hospital Tübingen, 72076 Tübingen, Germany
| | - Vijay P. S. Rawat
- Comprehensive Cancer Center Ulm, Institute of Experimental Cancer Research, University Hospital Ulm, 89081 Ulm, Germany; (L.M.K.); (V.P.S.R.)
- Special Centre for Molecular Medicine, Jawaharlal Nehru University, Delhi 110067, India
| | - Mirco Glitscher
- Department of Virology, Paul-Ehrlich-Institute, 63225 Langen, Germany; (M.G.); (E.H.)
| | - Eberhard Hildt
- Department of Virology, Paul-Ehrlich-Institute, 63225 Langen, Germany; (M.G.); (E.H.)
| | - Daniel Tews
- Department of Pediatrics and Adolescent Medicine, University Hospital Ulm, 89081 Ulm, Germany;
| | - Zachary Hunter
- Bing Center for Waldenström’s Macroglobulinemia, Boston, MA 02215, USA;
| | - Jan Münch
- Institute of Molecular Virology, Ulm University Medical Center, 89081 Ulm, Germany; (M.H.); (D.S.); (J.M.)
| | - Christian Buske
- Comprehensive Cancer Center Ulm, Institute of Experimental Cancer Research, University Hospital Ulm, 89081 Ulm, Germany; (L.M.K.); (V.P.S.R.)
- Department of Internal Medicine III, University Hospital Ulm, 89081 Ulm, Germany
| |
Collapse
|
36
|
Neidleman J, Luo X, George AF, McGregor M, Yang J, Yun C, Murray V, Gill G, Greene WC, Vasquez J, Lee S, Ghosn E, Lynch K, Roan NR. Distinctive features of SARS-CoV-2-specific T cells predict recovery from severe COVID-19. MEDRXIV : THE PREPRINT SERVER FOR HEALTH SCIENCES 2021:2021.01.22.21250054. [PMID: 33532792 PMCID: PMC7852243 DOI: 10.1101/2021.01.22.21250054] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 12/29/2022]
Abstract
Although T cells are likely players in SARS-CoV-2 immunity, little is known about the phenotypic features of SARS-CoV-2-specific T cells associated with recovery from severe COVID-19. We analyzed T cells from longitudinal specimens of 34 COVID-19 patients with severities ranging from mild (outpatient) to critical culminating in death. Relative to patients that succumbed, individuals that recovered from severe COVID-19 harbored elevated and increasing numbers of SARS-CoV-2-specific T cells capable of homeostatic proliferation. In contrast, fatal COVID-19 displayed elevated numbers of SARS-CoV-2-specific regulatory T cells and a time-dependent escalation in activated bystander CXCR4+ T cells. Together with the demonstration of increased proportions of inflammatory CXCR4+ T cells in the lungs of severe COVID-19 patients, these results support a model whereby lung-homing T cells activated through bystander effects contribute to immunopathology, while a robust, non-suppressive SARS-CoV-2-specific T cell response limits pathogenesis and promotes recovery from severe COVID-19.
Collapse
Affiliation(s)
- Jason Neidleman
- Gladstone Institutes, San Francisco, CA, USA
- Department of Urology, University of California, San Francisco, CA, USA
| | - Xiaoyu Luo
- Gladstone Institutes, San Francisco, CA, USA
| | - Ashley F. George
- Gladstone Institutes, San Francisco, CA, USA
- Department of Urology, University of California, San Francisco, CA, USA
| | - Matthew McGregor
- Gladstone Institutes, San Francisco, CA, USA
- Department of Urology, University of California, San Francisco, CA, USA
| | - Junkai Yang
- Department of Medicine, Lowance Center for Human Immunology, Emory Vaccine Center, Emory University, Atlanta, GA, USA
| | - Cassandra Yun
- Department of Laboratory Medicine, University of California, San Francisco, CA USA
| | - Victoria Murray
- Zuckerberg San Francisco General Hospital and the University of California, San Francisco, CA, USA
| | - Gurjot Gill
- Zuckerberg San Francisco General Hospital and the University of California, San Francisco, CA, USA
| | - Warner C. Greene
- Gladstone Institutes, San Francisco, CA, USA
- Department of Medicine, University of California, San Francisco, CA, USA
| | - Joshua Vasquez
- Department of Medicine, University of California, San Francisco, CA, USA
| | - Sulggi Lee
- Zuckerberg San Francisco General Hospital and the University of California, San Francisco, CA, USA
| | - Eliver Ghosn
- Department of Medicine, Lowance Center for Human Immunology, Emory Vaccine Center, Emory University, Atlanta, GA, USA
- Department of Pediatrics, Lowance Center for Human Immunology, Emory Vaccine Center, Emory University, Atlanta, GA, USA
| | - Kara Lynch
- Department of Laboratory Medicine, University of California, San Francisco, CA USA
| | - Nadia R. Roan
- Gladstone Institutes, San Francisco, CA, USA
- Department of Urology, University of California, San Francisco, CA, USA
| |
Collapse
|
37
|
Kaiser LM, Hunter ZR, Treon SP, Buske C. CXCR4 in Waldenström's Macroglobulinema: chances and challenges. Leukemia 2021; 35:333-345. [PMID: 33273682 PMCID: PMC7862063 DOI: 10.1038/s41375-020-01102-3] [Citation(s) in RCA: 57] [Impact Index Per Article: 14.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2020] [Revised: 10/09/2020] [Accepted: 11/17/2020] [Indexed: 02/06/2023]
Abstract
It is one of the major aims in cancer research to improve our understanding of the underlying mechanisms which initiate and maintain tumor growth and to translate these findings into novel clinical diagnostic and therapeutic concepts with the ultimate goal to improve patient care. One of the greater success stories in this respect has been Waldenström's Macroglobulinemia (WM), which is an incurable B-cell neoplasm characterized by serum monoclonal immunoglobulin M (IgM) and clonal lymphoplasmacytic cells infiltrating the bone marrow. Recent years have succeeded to describe the molecular landscape of WM in detail, highlighting two recurrently mutated genes, the MYD88 and the CXCR4 genes: MYD88 with an almost constant and recurrent point mutation present in over 90% of patients and CXCR4 with over 40 different mutations in the coding region affecting up to 40% of patients. Intriguingly, both mutations are activating mutations leading in the case of CXCR4 to an indelible activation and perpetual signaling of the chemokine receptor. These data have shed light on the essential role of CXCR4 in this disease and have paved the way to use these findings for predicting treatment response to the Bruton tyrosine kinase (BTK) inhibitor ibrutinib and novel therapeutic approaches in WM, which might be transferable to other related CXCR4 positive diseases. Well known for its central role in cancer progression and distribution, CXCR4 is highlighted in this review with regard to its biology, prognostic and predictive relevance and therapeutic implications in WM.
Collapse
Affiliation(s)
- Lisa Marie Kaiser
- Institute of Experimental Cancer Research, CCC and University Hospital Ulm, Germany, 89081, Ulm, Germany
| | - Zachary R Hunter
- Bing Center for Waldenström's Macroglobulinemia, Dana-Farber Cancer Institute, Harvard Medical School, Boston, MA, USA
| | - Steven P Treon
- Bing Center for Waldenström's Macroglobulinemia, Dana-Farber Cancer Institute, Harvard Medical School, Boston, MA, USA
| | - Christian Buske
- Institute of Experimental Cancer Research, CCC and University Hospital Ulm, Germany, 89081, Ulm, Germany.
| |
Collapse
|
38
|
López-Gil JC, Martin-Hijano L, Hermann PC, Sainz B. The CXCL12 Crossroads in Cancer Stem Cells and Their Niche. Cancers (Basel) 2021; 13:cancers13030469. [PMID: 33530455 PMCID: PMC7866198 DOI: 10.3390/cancers13030469] [Citation(s) in RCA: 30] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2020] [Revised: 01/17/2021] [Accepted: 01/19/2021] [Indexed: 12/16/2022] Open
Abstract
Simple Summary CXCL12 and its receptors have been extensively studied in cancer, including their influence on cancer stem cells (CSCs) and their niche. This intensive research has led to a better understanding of the crosstalk between CXCL12 and CSCs, which has aided in designing several drugs that are currently being tested in clinical trials. However, a comprehensive review has not been published to date. The aim of this review is to provide an overview on how CXCL12 axes are involved in the regulation and maintenance of CSCs, their presence and influence at different cellular levels within the CSC niche, and the current state-of-the-art of therapeutic approaches aimed to target the CXCL12 crossroads. Abstract Cancer stem cells (CSCs) are defined as a subpopulation of “stem”-like cells within the tumor with unique characteristics that allow them to maintain tumor growth, escape standard anti-tumor therapies and drive subsequent repopulation of the tumor. This is the result of their intrinsic “stem”-like features and the strong driving influence of the CSC niche, a subcompartment within the tumor microenvironment that includes a diverse group of cells focused on maintaining and supporting the CSC. CXCL12 is a chemokine that plays a crucial role in hematopoietic stem cell support and has been extensively reported to be involved in several cancer-related processes. In this review, we will provide the latest evidence about the interactions between CSC niche-derived CXCL12 and its receptors—CXCR4 and CXCR7—present on CSC populations across different tumor entities. The interactions facilitated by CXCL12/CXCR4/CXCR7 axes seem to be strongly linked to CSC “stem”-like features, tumor progression, and metastasis promotion. Altogether, this suggests a role for CXCL12 and its receptors in the maintenance of CSCs and the components of their niche. Moreover, we will also provide an update of the therapeutic options being currently tested to disrupt the CXCL12 axes in order to target, directly or indirectly, the CSC subpopulation.
Collapse
Affiliation(s)
- Juan Carlos López-Gil
- Department of Cancer Biology, Instituto de Investigaciones Biomédicas “Alberto Sols” (IIBM), CSIC-UAM, 28029 Madrid, Spain; (J.C.L.-G.); (L.M.-H.)
- Department of Biochemistry, Universidad Autónoma de Madrid (UAM), 28029 Madrid, Spain
- Chronic Diseases and Cancer, Area 3-Instituto Ramon y Cajal de Investigación Sanitaria (IRYCIS), 28029 Madrid, Spain
| | - Laura Martin-Hijano
- Department of Cancer Biology, Instituto de Investigaciones Biomédicas “Alberto Sols” (IIBM), CSIC-UAM, 28029 Madrid, Spain; (J.C.L.-G.); (L.M.-H.)
- Department of Biochemistry, Universidad Autónoma de Madrid (UAM), 28029 Madrid, Spain
- Chronic Diseases and Cancer, Area 3-Instituto Ramon y Cajal de Investigación Sanitaria (IRYCIS), 28029 Madrid, Spain
| | - Patrick C. Hermann
- Department of Internal Medicine I, Ulm University, 89081 Ulm, Germany
- Correspondence: (P.C.H.); (B.S.J.)
| | - Bruno Sainz
- Department of Cancer Biology, Instituto de Investigaciones Biomédicas “Alberto Sols” (IIBM), CSIC-UAM, 28029 Madrid, Spain; (J.C.L.-G.); (L.M.-H.)
- Department of Biochemistry, Universidad Autónoma de Madrid (UAM), 28029 Madrid, Spain
- Chronic Diseases and Cancer, Area 3-Instituto Ramon y Cajal de Investigación Sanitaria (IRYCIS), 28029 Madrid, Spain
- Correspondence: (P.C.H.); (B.S.J.)
| |
Collapse
|
39
|
Hayn M, Blötz A, Rodríguez A, Vidal S, Preising N, Ständker L, Wiese S, Stürzel CM, Harms M, Gross R, Jung C, Kiene M, Jacob T, Pöhlmann S, Forssmann WG, Münch J, Sparrer KMJ, Seuwen K, Hahn BH, Kirchhoff F. Natural cystatin C fragments inhibit GPR15-mediated HIV and SIV infection without interfering with GPR15L signaling. Proc Natl Acad Sci U S A 2021; 118:e2023776118. [PMID: 33431697 PMCID: PMC7826402 DOI: 10.1073/pnas.2023776118] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022] Open
Abstract
GPR15 is a G protein-coupled receptor (GPCR) proposed to play a role in mucosal immunity that also serves as a major entry cofactor for HIV-2 and simian immunodeficiency virus (SIV). To discover novel endogenous GPR15 ligands, we screened a hemofiltrate (HF)-derived peptide library for inhibitors of GPR15-mediated SIV infection. Our approach identified a C-terminal fragment of cystatin C (CysC95-146) that specifically inhibits GPR15-dependent HIV-1, HIV-2, and SIV infection. In contrast, GPR15L, the chemokine ligand of GPR15, failed to inhibit virus infection. We found that cystatin C fragments preventing GPR15-mediated viral entry do not interfere with GPR15L signaling and are generated by proteases activated at sites of inflammation. The antiretroviral activity of CysC95-146 was confirmed in primary CD4+ T cells and is conserved in simian hosts of SIV infection. Thus, we identified a potent endogenous inhibitor of GPR15-mediated HIV and SIV infection that does not interfere with the physiological function of this GPCR.
Collapse
Affiliation(s)
- Manuel Hayn
- Institute of Molecular Virology, Ulm University Medical Center, 89081 Ulm, Germany
| | - Andrea Blötz
- Institute of Molecular Virology, Ulm University Medical Center, 89081 Ulm, Germany
| | - Armando Rodríguez
- Core Facility Functional Peptidomics, Ulm University Medical Center, 89081 Ulm, Germany
- Core Unit Mass Spectrometry and Proteomics, Ulm University Medical Center, 89081 Ulm, Germany
- PHARIS Biotec GmbH, 30625 Hannover, Germany
| | - Solange Vidal
- Novartis Institutes for Biomedical Research, 4056 Basel, Switzerland
| | - Nico Preising
- Core Facility Functional Peptidomics, Ulm University Medical Center, 89081 Ulm, Germany
| | - Ludger Ständker
- Core Facility Functional Peptidomics, Ulm University Medical Center, 89081 Ulm, Germany
| | - Sebastian Wiese
- Core Unit Mass Spectrometry and Proteomics, Ulm University Medical Center, 89081 Ulm, Germany
| | - Christina M Stürzel
- Institute of Molecular Virology, Ulm University Medical Center, 89081 Ulm, Germany
| | - Mirja Harms
- Institute of Molecular Virology, Ulm University Medical Center, 89081 Ulm, Germany
| | - Rüdiger Gross
- Institute of Molecular Virology, Ulm University Medical Center, 89081 Ulm, Germany
| | - Christoph Jung
- Institute of Electrochemistry, Ulm University, 89081 Ulm, Germany
| | - Miriam Kiene
- Infection Biology Unit, German Primate Center-Leibniz Institute for Primate Research, 37077 Göttingen, Germany
| | - Timo Jacob
- Institute of Electrochemistry, Ulm University, 89081 Ulm, Germany
| | - Stefan Pöhlmann
- Infection Biology Unit, German Primate Center-Leibniz Institute for Primate Research, 37077 Göttingen, Germany
- Faculty of Biology and Psychology, University Göttingen, 37073 Göttingen, Germany
| | | | - Jan Münch
- Institute of Molecular Virology, Ulm University Medical Center, 89081 Ulm, Germany
| | | | - Klaus Seuwen
- Novartis Institutes for Biomedical Research, 4056 Basel, Switzerland
| | - Beatrice H Hahn
- Department of Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104-6076;
- Department of Microbiology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104-6076
| | - Frank Kirchhoff
- Institute of Molecular Virology, Ulm University Medical Center, 89081 Ulm, Germany;
| |
Collapse
|
40
|
Cadima-Couto I, Tauzin A, Freire JM, Figueira TN, Silva RDM, Pérez-Peinado C, Cunha-Santos C, Bártolo I, Taveira N, Gano L, Correia JDG, Goncalves J, Mammano F, Andreu D, Castanho MARB, Veiga AS. Anti-HIV-1 Activity of pepRF1, a Proteolysis-Resistant CXCR4 Antagonist Derived from Dengue Virus Capsid Protein. ACS Infect Dis 2021; 7:6-22. [PMID: 33319557 DOI: 10.1021/acsinfecdis.9b00507] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
Abstract
There is an urgent need for the development of new anti-HIV drugs that can complement existing medicines to be used against resistant strains. Here, we report the anti-HIV-1 peptide pepRF1, a human serum-resistant peptide derived from the Dengue virus capsid protein. In vitro, pepRF1 shows a 50% inhibitory concentration of 1.5 nM with a potential therapeutic window higher than 53 000. This peptide is specific for CXCR4-tropic strains, preventing viral entry into target cells by binding to the viral coreceptor CXCR4, acting as an antagonist of this receptor. pepRF1 is more effective than T20, the only peptide-based HIV-1 entry inhibitor approved, and excels in inhibiting a HIV-1 strain resistant to T20. Potentially, pepRF1 can be used alone or in combination with other anti-HIV drugs. Furthermore, one can also envisage its use as a novel therapeutic strategy for other CXCR4-related diseases.
Collapse
Affiliation(s)
- Iris Cadima-Couto
- Instituto de Medicina Molecular, Faculdade de Medicina, Universidade de Lisboa, Av. Prof. Egas Moniz, 1649-028 Lisboa, Portugal
| | - Alexandra Tauzin
- INSERM UMR 1124, Université de Paris, 45 rue des Saints Pères, F-75006 Paris, France
| | - João M. Freire
- Instituto de Medicina Molecular, Faculdade de Medicina, Universidade de Lisboa, Av. Prof. Egas Moniz, 1649-028 Lisboa, Portugal
| | - Tiago N. Figueira
- Instituto de Medicina Molecular, Faculdade de Medicina, Universidade de Lisboa, Av. Prof. Egas Moniz, 1649-028 Lisboa, Portugal
| | - Rúben D. M. Silva
- Centro de Ciências e Tecnologias Nucleares and Departamento de Engenharia e Ciências Nucleares, Instituto Superior Técnico, Universidade de Lisboa, CTN, Estrada Nacional 10 (km 139,7), 2695-066 Bobadela LRS, Portugal
| | - Clara Pérez-Peinado
- Department of Experimental and Health Sciences, Universitat Pompeu Fabra, Barcelona Biomedical Research Park, 08003 Barcelona, Spain
| | - Catarina Cunha-Santos
- Research Institute for Medicines (iMed.ULisboa), Faculdade de Farmácia, Universidade de Lisboa, Avenida Prof. Gama Pinto, 1649-003 Lisboa, Portugal
| | - Inês Bártolo
- Research Institute for Medicines (iMed.ULisboa), Faculdade de Farmácia, Universidade de Lisboa, Avenida Prof. Gama Pinto, 1649-003 Lisboa, Portugal
| | - Nuno Taveira
- Research Institute for Medicines (iMed.ULisboa), Faculdade de Farmácia, Universidade de Lisboa, Avenida Prof. Gama Pinto, 1649-003 Lisboa, Portugal
- Centro de Investigação Interdisciplinar Egas Moniz (CiiEM), Instituto Universitário Egas Moniz, 2829-511 Monte de Caparica, Portugal
| | - Lurdes Gano
- Centro de Ciências e Tecnologias Nucleares and Departamento de Engenharia e Ciências Nucleares, Instituto Superior Técnico, Universidade de Lisboa, CTN, Estrada Nacional 10 (km 139,7), 2695-066 Bobadela LRS, Portugal
| | - João D. G. Correia
- Centro de Ciências e Tecnologias Nucleares and Departamento de Engenharia e Ciências Nucleares, Instituto Superior Técnico, Universidade de Lisboa, CTN, Estrada Nacional 10 (km 139,7), 2695-066 Bobadela LRS, Portugal
| | - Joao Goncalves
- Research Institute for Medicines (iMed.ULisboa), Faculdade de Farmácia, Universidade de Lisboa, Avenida Prof. Gama Pinto, 1649-003 Lisboa, Portugal
| | - Fabrizio Mammano
- INSERM UMR 1124, Université de Paris, 45 rue des Saints Pères, F-75006 Paris, France
| | - David Andreu
- Department of Experimental and Health Sciences, Universitat Pompeu Fabra, Barcelona Biomedical Research Park, 08003 Barcelona, Spain
| | - Miguel A. R. B. Castanho
- Instituto de Medicina Molecular, Faculdade de Medicina, Universidade de Lisboa, Av. Prof. Egas Moniz, 1649-028 Lisboa, Portugal
| | - Ana Salomé Veiga
- Instituto de Medicina Molecular, Faculdade de Medicina, Universidade de Lisboa, Av. Prof. Egas Moniz, 1649-028 Lisboa, Portugal
| |
Collapse
|
41
|
Yang P, Hu Y, Zhou Q. The CXCL12-CXCR4 Signaling Axis Plays a Key Role in Cancer Metastasis and is a Potential Target for Developing Novel Therapeutics against Metastatic Cancer. Curr Med Chem 2020; 27:5543-5561. [PMID: 31724498 DOI: 10.2174/0929867326666191113113110] [Citation(s) in RCA: 22] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2018] [Revised: 10/07/2019] [Accepted: 10/25/2019] [Indexed: 12/12/2022]
Abstract
Metastasis is the main cause of death in cancer patients; there is currently no effective treatment for cancer metastasis. This is primarily due to our insufficient understanding of the metastatic mechanisms in cancer. An increasing number of studies have shown that the C-X-C motif chemokine Ligand 12 (CXCL12) is overexpressed in various tissues and organs. It is a key niche factor that nurtures the pre-metastatic niches (tumorigenic soil) and recruits tumor cells (oncogenic "seeds") to these niches, thereby fostering cancer cell aggression and metastatic capabilities. However, the C-X-C motif chemokine Receptor 4 (CXCR4) is aberrantly overexpressed in various cancer stem/progenitor cells and functions as a CXCL12 receptor. CXCL12 activates CXCR4 as well as multiple downstream multiple tumorigenic signaling pathways, promoting the expression of various oncogenes. Activation of the CXCL12-CXCR4 signaling axis promotes Epithelial-Mesenchymal Transition (EMT) and mobilization of cancer stem/progenitor cells to pre-metastatic niches. It also nurtures cancer cells with high motility, invasion, and dissemination phenotypes, thereby escalating multiple proximal or distal cancer metastasis; this results in poor patient prognosis. Based on this evidence, recent studies have explored either CXCL12- or CXCR4-targeted anti-cancer therapeutics and have achieved promising results in the preclinical trials. Further exploration of this new strategy and its potent therapeutics effect against metastatic cancer through the targeting of the CXCL12- CXCR4 signaling axis may lead to a novel therapy that can clean up the tumor microenvironment ("soil") and kill the cancer cells, particularly the cancer stem/progenitor cells ("seeds"), in cancer patients. Ultimately, this approach has the potential to effectively treat metastatic cancer.
Collapse
Affiliation(s)
- Ping Yang
- Department of Pathophysiology, School of Medicine (School of Nursing), Nantong University, Nantong, Jiangsu 226000, China
| | - Yae Hu
- Department of Pathophysiology, School of Medicine (School of Nursing), Nantong University, Nantong, Jiangsu 226000, China
| | - Quansheng Zhou
- Cyrus Tang Hematology Center, Jiangsu Institute of Hematology, Soochow University; Suzhou, Jiangsu 215123, China
| |
Collapse
|
42
|
Eiger DS, Boldizsar N, Honeycutt CC, Gardner J, Rajagopal S. Biased agonism at chemokine receptors. Cell Signal 2020; 78:109862. [PMID: 33249087 DOI: 10.1016/j.cellsig.2020.109862] [Citation(s) in RCA: 30] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2020] [Revised: 11/07/2020] [Accepted: 11/24/2020] [Indexed: 12/11/2022]
Abstract
In the human chemokine system, interactions between the approximately 50 known endogenous chemokine ligands and 20 known chemokine receptors (CKRs) regulate a wide range of cellular functions and biological processes including immune cell activation and homeostasis, development, angiogenesis, and neuromodulation. CKRs are a family of G protein-coupled receptors (GPCR), which represent the most common and versatile class of receptors in the human genome and the targets of approximately one third of all Food and Drug Administration-approved drugs. Chemokines and CKRs bind with significant promiscuity, as most CKRs can be activated by multiple chemokines and most chemokines can activate multiple CKRs. While these ligand-receptor interactions were previously regarded as redundant, it is now appreciated that many chemokine:CKR interactions display biased agonism, the phenomenon in which different ligands binding to the same receptor signal through different pathways with different efficacies, leading to distinct biological effects. Notably, these biased responses can be modulated through changes in ligand, receptor, and or the specific cellular context (system). In this review, we explore the biochemical mechanisms, functional consequences, and therapeutic potential of biased agonism in the chemokine system. An enhanced understanding of biased agonism in the chemokine system may prove transformative in the understanding of the mechanisms and consequences of biased signaling across all GPCR subtypes and aid in the development of biased pharmaceuticals with increased therapeutic efficacy and safer side effect profiles.
Collapse
Affiliation(s)
| | - Noelia Boldizsar
- Trinity College of Arts and Sciences, Duke University, Durham, NC 27710, USA.
| | | | - Julia Gardner
- Trinity College of Arts and Sciences, Duke University, Durham, NC 27710, USA.
| | - Sudarshan Rajagopal
- Department of Biochemistry, Duke University, Durham, NC 27710, USA; Department of Medicine, Duke University, Durham, NC 27710, USA.
| |
Collapse
|
43
|
Advanced fluorescence microscopy reveals disruption of dynamic CXCR4 dimerization by subpocket-specific inverse agonists. Proc Natl Acad Sci U S A 2020; 117:29144-29154. [PMID: 33148803 PMCID: PMC7682396 DOI: 10.1073/pnas.2013319117] [Citation(s) in RCA: 46] [Impact Index Per Article: 9.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
Class A G protein−coupled receptors (GPCRs) can form dimers and oligomers via poorly understood mechanisms. We show here that the chemokine receptor CXCR4, which is a major pharmacological target, has an oligomerization behavior modulated by its active conformation. Combining advanced, single-molecule, and single-cell optical tools with functional assays and computational approaches, we unveil three key features of CXCR4 quaternary organization: CXCR4 dimerization 1) is dynamic, 2) increases with receptor expression level, and 3) can be disrupted by stabilizing an inactive receptor conformation. Ligand binding motifs reveal a ligand binding subpocket essential to modulate both CXCR4 basal activity and dimerization. This is relevant to develop new strategies to design CXCR4-targeting drugs. Although class A G protein−coupled receptors (GPCRs) can function as monomers, many of them form dimers and oligomers, but the mechanisms and functional relevance of such oligomerization is ill understood. Here, we investigate this problem for the CXC chemokine receptor 4 (CXCR4), a GPCR that regulates immune and hematopoietic cell trafficking, and a major drug target in cancer therapy. We combine single-molecule microscopy and fluorescence fluctuation spectroscopy to investigate CXCR4 membrane organization in living cells at densities ranging from a few molecules to hundreds of molecules per square micrometer of the plasma membrane. We observe that CXCR4 forms dynamic, transient homodimers, and that the monomer−dimer equilibrium is governed by receptor density. CXCR4 inverse agonists that bind to the receptor minor pocket inhibit CXCR4 constitutive activity and abolish receptor dimerization. A mutation in the minor binding pocket reduced the dimer-disrupting ability of these ligands. In addition, mutating critical residues in the sixth transmembrane helix of CXCR4 markedly diminished both basal activity and dimerization, supporting the notion that CXCR4 basal activity is required for dimer formation. Together, these results link CXCR4 dimerization to its density and to its activity. They further suggest that inverse agonists binding to the minor pocket suppress both dimerization and constitutive activity and may represent a specific strategy to target CXCR4.
Collapse
|
44
|
Microtiter plate-based antibody-competition assay to determine binding affinities and plasma/blood stability of CXCR4 ligands. Sci Rep 2020; 10:16036. [PMID: 32994431 PMCID: PMC7525492 DOI: 10.1038/s41598-020-73012-4] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2020] [Accepted: 09/07/2020] [Indexed: 12/28/2022] Open
Abstract
C-X-C chemokine receptor type 4 (CXCR4) is involved in several intractable disease processes, including HIV infection, cancer cell metastasis, leukemia cell progression, rheumatoid arthritis, asthma and pulmonary fibrosis. Thus, CXCR4 represents a promising drug target and several CXCR4 antagonizing agents are in preclinical or clinical development. Important parameters in drug lead evaluation are determination of binding affinities to the receptor and assessment of their stability and activity in plasma or blood of animals and humans. Here, we designed a microtiter plate-based CXCR4 antibody competition assay that enables to measure inhibitory concentrations (IC50 values) and affinity constants (Ki values) of CXCR4 targeting drugs. The assay is based on the observation that most if not all CXCR4 antagonists compete with binding of the fluorescence-tagged CXCR4 antibody 12G5 to the receptor. We demonstrate that this antibody-competition assay allows a convenient and cheap determination of binding affinities of various CXCR4 antagonists in living cells within just 3 h. Moreover, the assay can be performed in the presence of high concentrations of physiologically relevant body fluids, and thus is a useful readout to evaluate stability (i.e. half-life) of CXCR4 ligands in serum/plasma, and even whole human and mouse blood ex vivo. Thus, this optimized 12G5 antibody-competition assay allows a robust and convenient determination and calculation of various important pharmacological parameters of CXCR4 receptor-drug interaction and may not only foster future drug development but also animal welfare by reducing the number of experimental animals.
Collapse
|
45
|
Hohloch K, Ziepert M, Truemper L, Buske C, Held G, Poeschel V, Chapuy B, Altmann B. Low serum albumin is an independent risk factor in elderly patients with aggressive B-cell lymphoma: Results from prospective trials of the German High-Grade Non-Hodgkin's Lymphoma Study Group. EJHAEM 2020; 1:181-187. [PMID: 35847697 PMCID: PMC9175786 DOI: 10.1002/jha2.61] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/22/2020] [Revised: 06/30/2020] [Accepted: 07/02/2020] [Indexed: 12/18/2022]
Abstract
Serum albumin a well-known risk factor predicting outcome in many solid tumors. We explore the role of low serum albumin (≤3.5 g/dL) as an independent risk factor in elderly patients with aggressive B-cell lymphoma. Outcome of 429 patients treated with R-CHOP-14 in the RICOVER-60 trial and available serum albumin were analyzed in this retrospective study. Of the 429 patients in the RICOVER-60 trial, 137 (32%) had low and 292 (68%) had normal serum albumin levels (>3.5 g/dL). In the low albumin group, patients had significantly higher International Prognostic Index (IPI), bulky disease, extralymphatic involvement, and B-symptoms. Event-free survival (EFS) (P < .001), progression-free survival (PFS) (P < .001), and overall survival (OS) (P < .001) were significantly inferior for patients with low compared to those with normal serum albumin. Multivariate analysis adjusted for IPI shows following Hazard ratios (HR) for low serum albumin: EFS (HR = 1.5; 95% confidance interval [CI] [1.1; 2.1], P = .009), PFS (HR = 1.7; 95% CI [1.2; 2.4], P = .001) and OS (HR = 1.6; 95% CI [1.1; 2.3], P = .006). Results were confirmed in 185 patients from the DENSE-R-CHOP-14 and SMARTE-R-CHOP-14 trials. In conclusion, low serum albumin is an independent risk factor in elderly patients with aggressive B-cell lymphoma treated with R-CHOP.
Collapse
Affiliation(s)
- Karin Hohloch
- Department of Hematology and OncologyKantonsspital GraubündenChurSwitzerland
- Department of Hematology and OncologyUniversity Medical CenterGeorg‐August UniversityGöttingenGermany
| | - Marita Ziepert
- Statistics and EpidemiologyInstitute for Medical InformaticsUniversity of LeipzigLeipzigGermany
| | - Lorenz Truemper
- Department of Hematology and OncologyUniversity Medical CenterGeorg‐August UniversityGöttingenGermany
| | - Christian Buske
- Comprehensive Cancer Center UlmInstitute of Experimental Cancer ResearchUniversity Hospital UlmUlmGermany
| | - Gerhard Held
- Department of Internal MedicineUniversity Hospital SaarlandHomburgGermany
| | - Viola Poeschel
- Department of Internal MedicineUniversity Hospital SaarlandHomburgGermany
| | - Bjoern Chapuy
- Department of Hematology and OncologyUniversity Medical CenterGeorg‐August UniversityGöttingenGermany
| | - Bettina Altmann
- Statistics and EpidemiologyInstitute for Medical InformaticsUniversity of LeipzigLeipzigGermany
| |
Collapse
|
46
|
Romero-Molina S, Ruiz-Blanco YB, Green JR, Sanchez-Garcia E. ProtDCal-Suite: A web server for the numerical codification and functional analysis of proteins. Protein Sci 2020; 28:1734-1743. [PMID: 31271472 DOI: 10.1002/pro.3673] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2019] [Revised: 06/21/2019] [Accepted: 06/24/2019] [Indexed: 12/24/2022]
Abstract
Computational tools for the analysis of protein data and the prediction of biological properties are essential in life sciences and biomedical research. Here, we introduce ProtDCal-Suite, a web server comprising a set of machine learning-based methods for studying proteins. The main module of ProtDCal-Suite is the ProtDCal software. ProtDCal translates the structural information of proteins into numerical descriptors that serve as input to machine-learning techniques. The ProtDCal-Suite server also incorporates a post-processing optional stage that allows ranking and filtering the obtained descriptors by computing their Shannon entropy values across the input set of proteins. ProtDCal's codification was used in the development of models for the prediction of specific protein properties. Thus, the other modules of ProtDCal-Suite are protein analysis tools implemented using ProtDCal's descriptors. Among them are PPI-Detect, for predicting the interaction likelihood of protein-protein and protein-peptide pairs, Enzyme Identifier, for identifying enzymes from amino acid sequences or 3D structures, and Pred-NGlyco, for predicting N-glycosylation sites. ProtDCal-Suite is freely accessible at https://protdcal.zmb.uni-due.de.
Collapse
Affiliation(s)
- Sandra Romero-Molina
- Computational Biochemistry, Center of Medical Biotechnology, University of Duisburg-Essen, Essen, Germany
| | - Yasser B Ruiz-Blanco
- Computational Biochemistry, Center of Medical Biotechnology, University of Duisburg-Essen, Essen, Germany
| | - James R Green
- Systems and Computer Engineering, Carleton University, Ottawa, Ontario, Canada
| | - Elsa Sanchez-Garcia
- Computational Biochemistry, Center of Medical Biotechnology, University of Duisburg-Essen, Essen, Germany
| |
Collapse
|
47
|
Groß R, Bauer R, Krüger F, Rücker-Braun E, Olari LR, Ständker L, Preising N, Rodríguez AA, Conzelmann C, Gerbl F, Sauter D, Kirchhoff F, Hagemann B, Gačanin J, Weil T, Ruiz-Blanco YB, Sanchez-Garcia E, Forssmann WG, Mankertz A, Santibanez S, Stenger S, Walther P, Wiese S, Spellerberg B, Münch J. A Placenta Derived C-Terminal Fragment of β-Hemoglobin With Combined Antibacterial and Antiviral Activity. Front Microbiol 2020; 11:508. [PMID: 32328038 PMCID: PMC7153485 DOI: 10.3389/fmicb.2020.00508] [Citation(s) in RCA: 21] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2019] [Accepted: 03/09/2020] [Indexed: 01/08/2023] Open
Abstract
The placenta acts as physical and immunological barrier against the transmission of viruses and bacteria from mother to fetus. However, the specific mechanisms by which the placenta protects the developing fetus from viral and bacterial pathogens are poorly understood. To identify placental peptides and small proteins protecting from viral and bacterial infections, we generated a peptide library from 10 kg placenta by chromatographic means. Screening the resulting 250 fractions against Herpes-Simplex-Virus 2 (HSV-2), which is rarely transmitted through the placenta, in a cell-based system identified two adjacent fractions with significant antiviral activity. Further rounds of chromatographic purification and anti-HSV-2 testing allowed to purify the bioactive peptide. Mass spectrometry revealed the presence of a 36-mer derived from the C-terminal region of the hemoglobin β subunit. The purified and corresponding chemically synthesized peptide, termed HBB(112–147), inhibited HSV-2 infection in a dose-dependent manner, with a mean IC50 in the median μg/ml range. Full-length hemoglobin tetramer had no antiviral activity. HBB(112–147) did not impair infectivity by direct targeting of the virions but prevented HSV-2 infection at the cell entry level. The peptide was inactive against Human Immunodeficiency Virus Type 1, Rubella and Zika virus infection, suggesting a specific anti-HSV-2 mechanism. Notably, HBB(112–147) has previously been identified as broad-spectrum antibacterial agent. It is abundant in placenta, reaching concentrations between 280 and 740 μg/ml, that are well sufficient to inhibit HSV-2 and prototype Gram-positive and -negative bacteria. We here additionally show, that HBB(112–147) also acts potently against Pseudomonas aeruginosa strains (including a multi-drug resistant strain) in a dose dependent manner, while full-length hemoglobin is inactive. Interestingly, the antibacterial activity of HBB(112–147) was increased under acidic conditions, a hallmark of infection and inflammatory conditions. Indeed, we found that HBB(112–147) is released from the hemoglobin precursor by Cathepsin D and Napsin A, acidic proteases highly expressed in placental and other tissues. We propose that upon viral or bacterial infection, the abundant hemoglobin precursor is proteolytically processed to release HBB(112–147), a broadly active antimicrobial innate immune defense peptide.
Collapse
Affiliation(s)
- Rüdiger Groß
- Institute of Molecular Virology, Ulm University Medical Center, Ulm, Germany
| | - Richard Bauer
- Institute of Medical Microbiology and Hygiene, Ulm University Medical Center, Ulm, Germany
| | - Franziska Krüger
- Institute of Molecular Virology, Ulm University Medical Center, Ulm, Germany
| | - Elke Rücker-Braun
- Department of Medicine I, University Hospital of Dresden, Dresden, Germany
| | - Lia-Raluca Olari
- Institute of Molecular Virology, Ulm University Medical Center, Ulm, Germany
| | - Ludger Ständker
- Core Facility Functional Peptidomics, Ulm University Medical Center, Ulm, Germany
| | - Nico Preising
- Core Facility Functional Peptidomics, Ulm University Medical Center, Ulm, Germany
| | - Armando A Rodríguez
- Core Facility Functional Peptidomics, Ulm University Medical Center, Ulm, Germany.,Core Unit of Mass Spectrometry and Proteomics, Ulm University, Ulm, Germany
| | - Carina Conzelmann
- Institute of Molecular Virology, Ulm University Medical Center, Ulm, Germany
| | - Fabian Gerbl
- Institute of Medical Microbiology and Hygiene, Ulm University Medical Center, Ulm, Germany
| | - Daniel Sauter
- Institute of Molecular Virology, Ulm University Medical Center, Ulm, Germany
| | - Frank Kirchhoff
- Institute of Molecular Virology, Ulm University Medical Center, Ulm, Germany
| | - Benjamin Hagemann
- Institute of Medical Microbiology and Hygiene, Ulm University Medical Center, Ulm, Germany
| | - Jasmina Gačanin
- Max Planck Institute for Polymer Research, Mainz, Germany.,Institute of Inorganic Chemistry I, University of Ulm, Ulm, Germany
| | - Tanja Weil
- Max Planck Institute for Polymer Research, Mainz, Germany.,Institute of Inorganic Chemistry I, University of Ulm, Ulm, Germany
| | - Yasser B Ruiz-Blanco
- Computational Biochemistry, Faculty of Biology, University of Duisburg-Essen, Essen, Germany
| | - Elsa Sanchez-Garcia
- Computational Biochemistry, Faculty of Biology, University of Duisburg-Essen, Essen, Germany
| | | | - Annette Mankertz
- WHO Measles/Rubella European RRL and NRC Measles, Mumps, Rubella, Robert Koch-Institute, Berlin, Germany
| | - Sabine Santibanez
- WHO Measles/Rubella European RRL and NRC Measles, Mumps, Rubella, Robert Koch-Institute, Berlin, Germany
| | - Steffen Stenger
- Institute of Medical Microbiology and Hygiene, Ulm University Medical Center, Ulm, Germany
| | - Paul Walther
- Central Facility for Electron Microscopy, Ulm University, Ulm, Germany
| | - Sebastian Wiese
- Core Unit of Mass Spectrometry and Proteomics, Ulm University, Ulm, Germany
| | - Barbara Spellerberg
- Institute of Medical Microbiology and Hygiene, Ulm University Medical Center, Ulm, Germany
| | - Jan Münch
- Institute of Molecular Virology, Ulm University Medical Center, Ulm, Germany.,Core Facility Functional Peptidomics, Ulm University Medical Center, Ulm, Germany
| |
Collapse
|
48
|
Al-Harthi S, Lachowicz JI, Nowakowski ME, Jaremko M, Jaremko Ł. Towards the functional high-resolution coordination chemistry of blood plasma human serum albumin. J Inorg Biochem 2019; 198:110716. [PMID: 31153112 DOI: 10.1016/j.jinorgbio.2019.110716] [Citation(s) in RCA: 88] [Impact Index Per Article: 14.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2019] [Revised: 05/07/2019] [Accepted: 05/13/2019] [Indexed: 12/11/2022]
Abstract
Human serum albumin (HSA) is a monomeric, globular, multi-carrier and the most abundant protein in the blood. HSA displays multiple ligand binding sites with extraordinary binding capacity for a wide range of ions and molecules. For decades, HSA's ability to bind to various ligands has led many scientists to study its physiological properties and protein structure; indeed, a better understanding of HSA-ligand interactions in human blood, at the atomic level, will likely foster the development of more potent, and overall more performant, diagnostic and therapeutic tools against serious human disorders such as diabetes, cardiovascular disorders, and cancer. Here, we present a concise overview of the current knowledge of HSA's structural characteristics, and its coordination chemistry with transition metal ions, within the scope and limitations of current techniques and biophysical methods to reach atomic resolution in solution and in blood serum. We also highlight the overwhelming need of a detailed atomistic understanding of HSA dynamic structures and interactions that are transient, weak, multi-site and multi-step, and allosterically affected by each other. Considering the fact that HSA is a current clinical tool for drug delivery systems and a potential contender as molecular cargo and nano-vehicle used in biophysical, clinical and industrial fields, we underline the emerging need for novel approaches to target the dynamic functional coordination chemistry of the human blood serum albumin in solution, at the atomic level.
Collapse
Affiliation(s)
- Samah Al-Harthi
- King Abdullah University of Science and Technology (KAUST), Biological and Environmental Science and Engineering Division (BESE), 23955-6900 Thuwal, Saudi Arabia
| | - Joanna Izabela Lachowicz
- Dipartimento di Scienze Chimiche e Geologiche, Università di Cagliari, Cittadella Universitaria, I-09042 Monserrato, Cagliari, Italy
| | - Michal Eligiusz Nowakowski
- King Abdullah University of Science and Technology (KAUST), Biological and Environmental Science and Engineering Division (BESE), 23955-6900 Thuwal, Saudi Arabia; Faculty of Chemistry, Biological and Chemical Research Centre, University of Warsaw, Żwirki i Wigury 101, 02-089 Warszawa, Poland
| | - Mariusz Jaremko
- King Abdullah University of Science and Technology (KAUST), Biological and Environmental Science and Engineering Division (BESE), 23955-6900 Thuwal, Saudi Arabia
| | - Łukasz Jaremko
- King Abdullah University of Science and Technology (KAUST), Biological and Environmental Science and Engineering Division (BESE), 23955-6900 Thuwal, Saudi Arabia.
| |
Collapse
|
49
|
Muratspahić E, Freissmuth M, Gruber CW. Nature-Derived Peptides: A Growing Niche for GPCR Ligand Discovery. Trends Pharmacol Sci 2019; 40:309-326. [PMID: 30955896 DOI: 10.1016/j.tips.2019.03.004] [Citation(s) in RCA: 48] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2018] [Revised: 02/26/2019] [Accepted: 03/06/2019] [Indexed: 12/11/2022]
Abstract
G protein-coupled receptors (GPCRs) represent important drug targets, as they regulate pivotal physiological processes and they have proved to be readily druggable. Natural products have been and continue to be amongst the most valuable sources for drug discovery and development. Here, we surveyed small molecules and (poly-)peptides derived from plants, animals, fungi, and bacteria, which modulate GPCR signaling. Among naturally occurring compounds, peptides from plants, cone-snails, snakes, spiders, scorpions, fungi, and bacteria are of particular interest as lead compounds for the development of GPCR ligands, since they cover a chemical space, which differs from that of synthetic small molecules. Peptides, however, face challenges, some of which can be overcome by studying plant-derived compounds. We argue here that the opportunities outweigh the challenges.
Collapse
Affiliation(s)
- Edin Muratspahić
- Institute of Pharmacology, Center for Physiology and Pharmacology, Medical University of Vienna, Austria
| | - Michael Freissmuth
- Institute of Pharmacology, Center for Physiology and Pharmacology, Medical University of Vienna, Austria; Gaston H. Glock Research Laboratories for Exploratory Drug Development, Center for Physiology and Pharmacology, Medical University of Vienna, Austria
| | - Christian W Gruber
- Institute of Pharmacology, Center for Physiology and Pharmacology, Medical University of Vienna, Austria.
| |
Collapse
|
50
|
Romero-Molina S, Ruiz-Blanco YB, Harms M, Münch J, Sanchez-Garcia E. PPI-Detect: A support vector machine model for sequence-based prediction of protein-protein interactions. J Comput Chem 2019; 40:1233-1242. [PMID: 30768790 DOI: 10.1002/jcc.25780] [Citation(s) in RCA: 44] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2018] [Revised: 11/29/2018] [Accepted: 12/29/2018] [Indexed: 12/18/2022]
Abstract
The prediction of peptide-protein or protein-protein interactions (PPI) is a challenging task, especially if amino acid sequences are the only information available. Machine learning methods allow us to exploit the information content in PPI datasets. However, the numerical codification of these datasets often influences the performance of data mining approaches. Here, we introduce a procedure for the general-purpose numerical codification of polypeptides. This procedure transforms pairs of amino acid sequences into a machine learning-friendly vector, whose elements represent numerical descriptors of residues in proteins. We used this numerical encoding procedure for the development of a support vector machine model (PPI-Detect), which allows predicting whether two proteins will interact or not. PPI-Detect (https://ppi-detect.zmb.uni-due.de/) outperforms state of the art sequence-based predictors of PPI. We employed PPI-Detect for the analysis of derivatives of EPI-X4, an endogenous peptide inhibitor of CXCR4, a G-protein-coupled receptor. There, we identified with high accuracy those peptides which bind better than EPI-X4 to the receptor. Also using PPI-Detect, we designed a novel peptide and then experimentally established its anti-CXCR4 activity. © 2019 Wiley Periodicals, Inc.
Collapse
Affiliation(s)
- Sandra Romero-Molina
- Center of Medical Biotechnology, University of Duisburg-Essen, Duisburg, Germany
| | - Yasser B Ruiz-Blanco
- Center of Medical Biotechnology, University of Duisburg-Essen, Duisburg, Germany
| | - Mirja Harms
- Institute of Molecular Virology, Ulm University Medical Center, Ulm, Germany
| | - Jan Münch
- Institute of Molecular Virology, Ulm University Medical Center, Ulm, Germany.,Core Facility Functional Peptidomics, Ulm University Medical Center, Ulm, Germany
| | - Elsa Sanchez-Garcia
- Center of Medical Biotechnology, University of Duisburg-Essen, Duisburg, Germany
| |
Collapse
|