1
|
Kokot I, Mazurek S, Piwowar A, Sokolik R, Rodak K, Kacperczyk M, Szostak R, Cuprych P, Korman L, Maria Kratz E. Comparative profiling of serum biomarkers and ATR-FTIR spectroscopy for differential diagnosis of patients with rheumatoid and psoriatic arthritis - a pilot study. SPECTROCHIMICA ACTA. PART A, MOLECULAR AND BIOMOLECULAR SPECTROSCOPY 2024; 321:124654. [PMID: 38941757 DOI: 10.1016/j.saa.2024.124654] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/05/2024] [Revised: 05/28/2024] [Accepted: 06/11/2024] [Indexed: 06/30/2024]
Abstract
BACKGROUND Rheumatoid arthritis (RA) and psoriatic arthritis (PsA) are chronic inflammatory diseases in which innate and adaptive responses of the immune system are induced. RA and PsA have complex signaling pathways. Despite the differences in their clinical presentation, there is a great demand for fast and accurate diagnosis of diseases to implement treatment and plan an individual therapeutic strategy quickly. In this report, we present the results of differential diagnosis of patients with RA and PsA and healthy subjects (C, control group), allowing for reliable differentiation of groups of rheumatoid patients based on biochemical parameters, attenuated total reflection Fourier-transform infrared (ATR-FTIR) spectra, and combined data sets. MATERIALS AND METHODS Biochemical analyses, ELISA (enzyme-linked immunosorbent assays), and multiplex assays were conducted for blood sera from patients with RA (n = 32), patients with PsA (n = 28), and the control group (n = 18). ATR-FTIR spectra were collected for lyophilized sera. RESULTS The combination of six biochemical parameters (WBC, ESR, RF, CRP, HCC-4/CCL16, and HMGB1/HMGB) allowed the development of the partial least squares discriminant analysis (PLS-DA) model with an overall accuracy (OA) of 80% for test samples. The best separation between RA, PsA, and the control group was obtained utilizing spectral data. Using the interval PLS algorithm (iPLS) specific spectral ranges were selected and a classifier characterized by OA value for test set equal to 88% was obtained. This parameter, for the hybrid PLS-DA model constructed using selected biochemical parameters and a significantly reduced number of spectral variables, reached the level of 84%. CONCLUSIONS PLS-DA models developed on the basis of spectral data enable effective differentiation of patients with RA, patients with PsA, and healthy subjects. They appeared to be insensitive to existing inflammation processes which opens interesting perspectives for new diagnostic tests and algorithms for identification of patients with RA and PsA.
Collapse
Affiliation(s)
- Izabela Kokot
- Department of Laboratory Diagnostics, Division of Laboratory Diagnostics, Faculty of Pharmacy, Wroclaw Medical University, Borowska Street 211A, 50-556 Wroclaw, Poland.
| | - Sylwester Mazurek
- Department of Chemistry, University of Wroclaw, F. Joliot-Curie 14, 50-383 Wroclaw, Poland.
| | - Agnieszka Piwowar
- Department of Toxicology, Faculty of Pharmacy, Wroclaw Medical University, Borowska Street 211, 50-556 Wroclaw, Poland
| | - Renata Sokolik
- Department of Rheumatology and Internal Medicine, Faculty of Medicine, Wroclaw Medical University, Borowska Street 213, 50-556 Wroclaw, Poland
| | - Kamil Rodak
- Department of Laboratory Diagnostics, Division of Laboratory Diagnostics, Faculty of Pharmacy, Wroclaw Medical University, Borowska Street 211A, 50-556 Wroclaw, Poland
| | - Monika Kacperczyk
- Department of Laboratory Diagnostics, Division of Laboratory Diagnostics, Faculty of Pharmacy, Wroclaw Medical University, Borowska Street 211A, 50-556 Wroclaw, Poland
| | - Roman Szostak
- Department of Chemistry, University of Wroclaw, F. Joliot-Curie 14, 50-383 Wroclaw, Poland
| | - Przemysław Cuprych
- Department of Chemistry, University of Wroclaw, F. Joliot-Curie 14, 50-383 Wroclaw, Poland
| | - Lucyna Korman
- Department of Rheumatology and Internal Medicine, Faculty of Medicine, Wroclaw Medical University, Borowska Street 213, 50-556 Wroclaw, Poland
| | - Ewa Maria Kratz
- Department of Laboratory Diagnostics, Division of Laboratory Diagnostics, Faculty of Pharmacy, Wroclaw Medical University, Borowska Street 211A, 50-556 Wroclaw, Poland
| |
Collapse
|
2
|
Shirian J, Hockla A, Gleba JJ, Coban M, Rotenberg N, Strik LM, Alasonyalilar Demirer A, Pawlush ML, Copland JA, Radisky ES, Shifman JM. Improving Circulation Half-Life of Therapeutic Candidate N-TIMP2 by Unfolded Peptide Extension. Biomolecules 2024; 14:1187. [PMID: 39334953 PMCID: PMC11429640 DOI: 10.3390/biom14091187] [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: 07/02/2024] [Revised: 09/16/2024] [Accepted: 09/17/2024] [Indexed: 09/30/2024] Open
Abstract
Matrix metalloproteinases (MMPs) are significant drivers of many diseases, including cancer, and are established targets for drug development. Tissue inhibitors of metalloproteinases (TIMPs) are endogenous MMP inhibitors and are being pursued for the development of anti-MMP therapeutics. TIMPs possess many attractive properties for drug candidates, such as complete MMP inhibition, low toxicity, low immunogenicity, and high tissue permeability. However, a major challenge with TIMPs is their rapid clearance from the bloodstream due to their small size. This study explores a method for extending the plasma half-life of the N-terminal domain of TIMP2 (N-TIMP2) by appending it with a long, intrinsically unfolded tail containing Pro, Ala, and Thr (PATylation). We designed and produced two PATylated N-TIMP2 constructs with tail lengths of 100 and 200 amino acids (N-TIMP2-PAT100 and N-TIMP2-PAT200). Both constructs demonstrated higher apparent molecular weights and retained high inhibitory activity against MMP-9. N-TIMP2-PAT200 significantly increased plasma half-life in mice compared to the non-PATylated variant, enhancing its therapeutic potential. PATylation offers distinct advantages for half-life extension, such as fully genetic encoding, monodispersion, and biodegradability. It can be easily applied to N-TIMP2 variants engineered for high affinity and selectivity toward individual MMPs, creating promising candidates for drug development against MMP-related diseases.
Collapse
Affiliation(s)
- Jason Shirian
- Department of Biological Chemistry, The Alexander Silberman Institute of Life Sciences, The Hebrew University of Jerusalem, Jerusalem 9190401, Israel
| | - Alexandra Hockla
- Department of Cancer Biology, Mayo Clinic Comprehensive Cancer Center, Jacksonville, FL 32224, USA (M.C.); (M.L.P.)
| | - Justyna J. Gleba
- Department of Cancer Biology, Mayo Clinic Comprehensive Cancer Center, Jacksonville, FL 32224, USA (M.C.); (M.L.P.)
| | - Matt Coban
- Department of Cancer Biology, Mayo Clinic Comprehensive Cancer Center, Jacksonville, FL 32224, USA (M.C.); (M.L.P.)
| | - Naama Rotenberg
- Department of Biological Chemistry, The Alexander Silberman Institute of Life Sciences, The Hebrew University of Jerusalem, Jerusalem 9190401, Israel
| | - Laura M. Strik
- Department of Cancer Biology, Mayo Clinic Comprehensive Cancer Center, Jacksonville, FL 32224, USA (M.C.); (M.L.P.)
| | - Aylin Alasonyalilar Demirer
- Department of Cancer Biology, Mayo Clinic Comprehensive Cancer Center, Jacksonville, FL 32224, USA (M.C.); (M.L.P.)
| | - Matt L. Pawlush
- Department of Cancer Biology, Mayo Clinic Comprehensive Cancer Center, Jacksonville, FL 32224, USA (M.C.); (M.L.P.)
| | - John A. Copland
- Department of Cancer Biology, Mayo Clinic Comprehensive Cancer Center, Jacksonville, FL 32224, USA (M.C.); (M.L.P.)
| | - Evette S. Radisky
- Department of Cancer Biology, Mayo Clinic Comprehensive Cancer Center, Jacksonville, FL 32224, USA (M.C.); (M.L.P.)
| | - Julia M. Shifman
- Department of Biological Chemistry, The Alexander Silberman Institute of Life Sciences, The Hebrew University of Jerusalem, Jerusalem 9190401, Israel
| |
Collapse
|
3
|
Shirian J, Hockla A, Gleba JJ, Coban M, Rotenberg N, Strik LM, Alasonyalilar Demirer A, Pawlush ML, Copland JA, Radisky ES, Shifman JM. Improving Circulation Half-Life of Therapeutic Candidate N-TIMP2 by Unfolded Peptide Extension. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.06.27.600979. [PMID: 38979353 PMCID: PMC11230438 DOI: 10.1101/2024.06.27.600979] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 07/10/2024]
Abstract
Matrix Metalloproteinases (MMPs) are drivers of many diseases including cancer and are established targets for drug development. Tissue inhibitors of metalloproteinases (TIMPs) are human proteins that inhibit MMPs and are being pursued for the development of anti-MMP therapeutics. TIMPs possess many attractive properties of a drug candidate, such as complete MMP inhibition, low toxicity and immunogenicity, high tissue permeability and others. A major challenge with TIMPs, however, is their formulation and delivery, as these proteins are quickly cleared from the bloodstream due to their small size. In this study, we explore a new method for plasma half-life extension for the N-terminal domain of TIMP2 (N-TIMP2) through appending it with a long intrinsically unfolded tail containing a random combination of Pro, Ala, and Thr (PATylation). We design, produce and explore two PATylated N-TIMP2 constructs with a tail length of 100- and 200-amino acids (N-TIMP2-PAT100 and N-TIMP2-PAT200, respectively). We demonstrate that both PATylated N-TIMP2 constructs possess apparent higher molecular weights compared to the wild-type protein and retain high inhibitory activity against MMP-9. Furthermore, when injected into mice, N-TIMP2-PAT200 exhibited a significant increase in plasma half-life compared to the non-PATylated variant, enhancing the therapeutic potential of the protein. Thus, we establish that PATylation could be successfully applied to TIMP-based therapeutics and offers distinct advantages as an approach for half-life extension, such as fully genetic encoding of the gene construct, mono-dispersion, and biodegradability. Furthermore, PATylation could be easily applied to N-TIMP2 variants engineered to possess high affinity and selectivity toward individual MMP family members, thus creating attractive candidates for drug development against MMP-related diseases.
Collapse
Affiliation(s)
- Jason Shirian
- Department of Biological Chemistry, The Alexander Silberman Institute of Life Sciences, The Hebrew University of Jerusalem, Jerusalem, 9190401, Israel
| | - Alexandra Hockla
- Department of Cancer Biology, Mayo Clinic Comprehensive Cancer Center, Jacksonville, Florida 32224, United States
| | - Justyna J. Gleba
- Department of Cancer Biology, Mayo Clinic Comprehensive Cancer Center, Jacksonville, Florida 32224, United States
| | - Matt Coban
- Department of Cancer Biology, Mayo Clinic Comprehensive Cancer Center, Jacksonville, Florida 32224, United States
| | - Naama Rotenberg
- Department of Biological Chemistry, The Alexander Silberman Institute of Life Sciences, The Hebrew University of Jerusalem, Jerusalem, 9190401, Israel
| | - Laura M. Strik
- Department of Cancer Biology, Mayo Clinic Comprehensive Cancer Center, Jacksonville, Florida 32224, United States
| | - Aylin Alasonyalilar Demirer
- Department of Cancer Biology, Mayo Clinic Comprehensive Cancer Center, Jacksonville, Florida 32224, United States
| | - Matt L. Pawlush
- Department of Cancer Biology, Mayo Clinic Comprehensive Cancer Center, Jacksonville, Florida 32224, United States
| | - John A. Copland
- Department of Cancer Biology, Mayo Clinic Comprehensive Cancer Center, Jacksonville, Florida 32224, United States
| | - Evette S. Radisky
- Department of Cancer Biology, Mayo Clinic Comprehensive Cancer Center, Jacksonville, Florida 32224, United States
| | - Julia M. Shifman
- Department of Biological Chemistry, The Alexander Silberman Institute of Life Sciences, The Hebrew University of Jerusalem, Jerusalem, 9190401, Israel
| |
Collapse
|
4
|
Taheri E, Raeeszadeh-Sarmazdeh M. Effect of TIMPs and Their Minimally Engineered Variants in Blocking Invasion and Migration of Brain Cancer Cells. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.06.05.597644. [PMID: 38895489 PMCID: PMC11185677 DOI: 10.1101/2024.06.05.597644] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/21/2024]
Abstract
Matrix metalloproteinases (MMPs) play a pivotal role in extracellular matrix (ECM) remodeling, influencing various aspects of cancer progression including migration, invasion, angiogenesis, and metastasis. Overexpression of MMPs, particularly MMP-2 and MMP-9, is notably pronounced in glioblastoma multiforme (GBM), a highly aggressive primary brain tumor characterized by diffuse and infiltrative behavior. Previous attempts to develop small molecule MMP inhibitors have failed in clinical trials, necessitating the exploration of more stable and selective alternatives. Tissue inhibitors of metalloproteinases (TIMPs), endogenous human proteins, offer promising potential due to their stability and broader interaction interfaces compared to small molecule inhibitors. In this study, we examined the effectiveness of wild-type human TIMP-1 and TIMP-3, alongside engineered minimal TIMP variants (mTC1 and mTC3), specifically designed for targeted MMP inhibition to reduce the migratory and invasive capabilities of GBM cells. Our investigation focused on these minimal TIMP variants, which provide enhanced tissue penetration and cellular uptake due to their small molecular weight, aiming to validate their potential as therapeutic agents. The results demonstrated that mTC1 and mTC3 effectively inhibit MMP activity, a critical factor in GBM aggressiveness, thereby highlighting their promise in controlling tumor spread. Given the lethality of GBM and the limited effectiveness of current treatments, the application of engineered TIMP variants represents a novel and potentially transformative therapeutic approach. By offering targeted MMP inhibition, these variants may significantly improve patient outcomes, providing new avenues for treatment and enhancing the survival and quality of life for patients with this devastating disease.
Collapse
|
5
|
Radisky ES. Extracellular proteolysis in cancer: Proteases, substrates, and mechanisms in tumor progression and metastasis. J Biol Chem 2024; 300:107347. [PMID: 38718867 PMCID: PMC11170211 DOI: 10.1016/j.jbc.2024.107347] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2024] [Revised: 04/08/2024] [Accepted: 04/25/2024] [Indexed: 06/02/2024] Open
Abstract
A vast ensemble of extracellular proteins influences the development and progression of cancer, shaped and reshaped by a complex network of extracellular proteases. These proteases, belonging to the distinct classes of metalloproteases, serine proteases, cysteine proteases, and aspartic proteases, play a critical role in cancer. They often become dysregulated in cancer, with increases in pathological protease activity frequently driven by the loss of normal latency controls, diminished regulation by endogenous protease inhibitors, and changes in localization. Dysregulated proteases accelerate tumor progression and metastasis by degrading protein barriers within the extracellular matrix (ECM), stimulating tumor growth, reactivating dormant tumor cells, facilitating tumor cell escape from immune surveillance, and shifting stromal cells toward cancer-promoting behaviors through the precise proteolysis of specific substrates to alter their functions. These crucial substrates include ECM proteins and proteoglycans, soluble proteins secreted by tumor and stromal cells, and extracellular domains of cell surface proteins, including membrane receptors and adhesion proteins. The complexity of the extracellular protease web presents a significant challenge to untangle. Nevertheless, technological strides in proteomics, chemical biology, and the development of new probes and reagents are enabling progress and advancing our understanding of the pivotal importance of extracellular proteolysis in cancer.
Collapse
Affiliation(s)
- Evette S Radisky
- Department of Cancer Biology, Mayo Clinic Comprehensive Cancer Center, Jacksonville, Florida, USA.
| |
Collapse
|
6
|
Kalantar M, Hilpert GA, Mosca ER, Raeeszadeh-Sarmazdeh M. Engineering metalloproteinase inhibitors: tissue inhibitors of metalloproteinases or antibodies, that is the question. Curr Opin Biotechnol 2024; 86:103094. [PMID: 38430575 DOI: 10.1016/j.copbio.2024.103094] [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: 12/18/2023] [Revised: 01/27/2024] [Accepted: 02/01/2024] [Indexed: 03/04/2024]
Abstract
Targeting metalloproteinases (MPs) has been the center of attention for developing therapeutics due to their contribution to a wide range of diseases, including cancer, cardiovascular, neurodegenerative disease, and preterm labor. Protein-based MP inhibitors offer higher stability and selectivity, which is critical for developing efficient therapeutics with low off-target effects. Tissue inhibitors of metalloproteinases (TIMPs), natural inhibitors of MPs, and antibodies provide excellent protein scaffolds for engineering selective or multispecific MP inhibitors. Advances in protein engineering and design techniques, such as rational design and directed evolution using yeast display to develop potent MP inhibitors, are discussed, including but not limited to loop grafting, swapping, and counterselective selection.
Collapse
Affiliation(s)
- Masoud Kalantar
- Chemical and Materials Engineering, University of Nevada, Reno, NV 89557, USA
| | - Gregory A Hilpert
- Chemical and Materials Engineering, University of Nevada, Reno, NV 89557, USA
| | - Ethan R Mosca
- Chemical and Materials Engineering, University of Nevada, Reno, NV 89557, USA
| | | |
Collapse
|
7
|
Ahmadighadykolaei H, Radisky ES, Raeeszadeh-Sarmazdeh M. Engineering Selective TIMPs Using a Counter-Selective Screening Strategy. Methods Mol Biol 2024; 2747:257-278. [PMID: 38038946 PMCID: PMC11235094 DOI: 10.1007/978-1-0716-3589-6_20] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/02/2023]
Abstract
The yeast surface display platform provides a powerful approach for screening protein diversity libraries to identify binders with an enhanced affinity toward a binding partner. Here, we describe an adaptation of the approach to identify binders with enhanced specificity toward one among multiple closely related binding partners. Specifically, we describe methods for engineering selective matrix metalloproteinase (MMP) inhibitors via yeast surface display of a tissue inhibitor of metalloproteinase (TIMP) diversity library coupled with a counter-selective screening strategy. This protocol may also be employed for developing selective protein binders or inhibitors toward other targets.
Collapse
|
8
|
Hosseini A, Kumar S, Hedin K, Raeeszadeh‐Sarmazdeh M. Engineering minimal tissue inhibitors of metalloproteinase targeting MMPs via gene shuffling and yeast surface display. Protein Sci 2023; 32:e4795. [PMID: 37807423 PMCID: PMC10659938 DOI: 10.1002/pro.4795] [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: 05/02/2023] [Revised: 09/21/2023] [Accepted: 09/23/2023] [Indexed: 10/10/2023]
Abstract
Overexpression of specific matrix metalloproteinases (MMPs) has a key role in development of several diseases, such as cancer, neurological disorders, and cardiovascular diseases due to their critical role in degradation and remodeling of the extracellular matrix (ECM). Tissue inhibitors of metalloproteinases (TIMPs), a family of four in humans, are endogenous inhibitors of MMPs. TIMPs have a high level of sequence and structure homology, with a broad range of binding and inhibition to the family of MMPs. It is important to identify the key motifs of TIMPs responsible for inhibition of MMPs to develop efficient therapeutics targeting specific MMPs. We used DNA shuffling between the human TIMP family to generate a minimal TIMP hybrid library in yeast to identify the dominant minimal MMP inhibitory regions. The minimal TIMP variants screened toward MMP-3 and MMP-9 using fluorescent-activated cell sorting (FACS). Interestingly, several minimal TIMP variants selected after screening toward MMP-3cd or MMP-9cd, with lengths as short as 20 amino acids, maintained or improved binding to MMP-3 and MMP-9. The TIMP-MMP binding dissociation constant (KD ), in the nM range, and MMP inhibition constants (Ki ), in the pM range, of these minimal TIMP variants were similar to the N-terminal domain of TIMP-1 on the yeast surface and in solution indicating the potency of these minimal variants as MMP inhibitors. We further used molecular modeling simulation, and molecular docking of the minimal TIMP variants in complex with MMP-3cd to understand the binding and inhibition mechanism of these variants.
Collapse
Affiliation(s)
- Arman Hosseini
- Department of Chemical and Materials EngineeringUniversity of NevadaRenoNVUSA
| | - Sachin Kumar
- Department of Chemical and Materials EngineeringUniversity of NevadaRenoNVUSA
| | - Kyle Hedin
- Department of Chemical and Materials EngineeringUniversity of NevadaRenoNVUSA
| | | |
Collapse
|
9
|
Shoari A, Khalili-Tanha G, Coban MA, Radisky ES. Structure and computation-guided yeast surface display for the evolution of TIMP-based matrix metalloproteinase inhibitors. Front Mol Biosci 2023; 10:1321956. [PMID: 38074088 PMCID: PMC10702220 DOI: 10.3389/fmolb.2023.1321956] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2023] [Accepted: 11/13/2023] [Indexed: 01/03/2024] Open
Abstract
The study of protein-protein interactions (PPIs) and the engineering of protein-based inhibitors often employ two distinct strategies. One approach leverages the power of combinatorial libraries, displaying large ensembles of mutant proteins, for example, on the yeast cell surface, to select binders. Another approach harnesses computational modeling, sifting through an astronomically large number of protein sequences and attempting to predict the impact of mutations on PPI binding energy. Individually, each approach has inherent limitations, but when combined, they generate superior outcomes across diverse protein engineering endeavors. This synergistic integration of approaches aids in identifying novel binders and inhibitors, fine-tuning specificity and affinity for known binding partners, and detailed mapping of binding epitopes. It can also provide insight into the specificity profiles of varied PPIs. Here, we outline strategies for directing the evolution of tissue inhibitors of metalloproteinases (TIMPs), which act as natural inhibitors of matrix metalloproteinases (MMPs). We highlight examples wherein design of combinatorial TIMP libraries using structural and computational insights and screening these libraries of variants using yeast surface display (YSD), has successfully optimized for MMP binding and selectivity, and conferred insight into the PPIs involved.
Collapse
Affiliation(s)
| | | | | | - Evette S. Radisky
- Department of Cancer Biology, Mayo Clinic, Jacksonville, FL, United States
| |
Collapse
|
10
|
Ahmadighadykolaei H, Lambert JA, Raeeszadeh-Sarmazdeh M. TIMP-1 Protects Tight Junctions of Brain Endothelial Cells From MMP-Mediated Degradation. Pharm Res 2023; 40:2121-2131. [PMID: 37700105 PMCID: PMC10878538 DOI: 10.1007/s11095-023-03593-y] [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: 06/08/2023] [Accepted: 08/18/2023] [Indexed: 09/14/2023]
Abstract
OBJECTIVE The blood-brain barrier (BBB) plays a critical role in central nervous system homeostasis, and the integrity of BBB is disrupted in many neurodegenerative diseases. Matrix metalloproteinases (MMPs) degrade the tight junctions (TJs) of endothelial cells and basement membrane components essential to BBB integrity, which leads to increased BBB permeability and allows inflammatory cells and neurotoxic substances to enter the brain. Tissue inhibitors of metalloproteinases (TIMPs), endogenous inhibitors of MMPs, regulate MMP activity, thereby maintaining BBB integrity. METHODS The disruptive impacts of MMP-3 and MMP-9 on BBB and protective effect of TIMP-1 were investigated in a simplified in vitro model of the BBB, which was generated using rat brain microvascular endothelial cells (RBMEC). The main features of BBB formation, including permeability and the trans-endothelial electrical resistance (TEER), were monitored over time after the addition of MMP-3 and MMP-9 and their complexes with TIMP-1 inhibitor. RESULTS Our results indicated that MMP-3 and MMP-9 caused a dose-dependent disruption of the BBB, with 1.5 µM MMPs resulting in an over threefold increase in permeability, while TIMP-1 inhibition protected the integrity of the BBB model and recovered TEER and permeability of RBMECs. The disruption and recovery of tight junction proteins of RBMECs after MMP and TIMP treatment were also detected using fluorescent microscopy. CONCLUSION MMP-9 and MMP-3 disrupt the BBB by degrading tight junctions in endothelial cells, and TIMP-1 could inhibit the disruptive effect of MMP-3 and MMP-9 by showing potential as therapeutic protein against MMP-related diseases where BBB disruption plays a role.
Collapse
Affiliation(s)
- Hannaneh Ahmadighadykolaei
- Department of Chemical and Materials Engineering, University of Nevada, 1664 N. Virginia St, Reno, NV, 89557, USA
| | - Janet A Lambert
- Department of Chemical and Materials Engineering, University of Nevada, 1664 N. Virginia St, Reno, NV, 89557, USA
- Department of Pharmacology, University of Nevada, Reno School of Medicine, Reno, NV, USA
| | - Maryam Raeeszadeh-Sarmazdeh
- Department of Chemical and Materials Engineering, University of Nevada, 1664 N. Virginia St, Reno, NV, 89557, USA.
| |
Collapse
|
11
|
Bonadio A, Wenig BL, Hockla A, Radisky ES, Shifman JM. Designed Loop Extension Followed by Combinatorial Screening Confers High Specificity to a Broad Matrix MetalloproteinaseInhibitor. J Mol Biol 2023; 435:168095. [PMID: 37068580 PMCID: PMC10312305 DOI: 10.1016/j.jmb.2023.168095] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2023] [Revised: 04/03/2023] [Accepted: 04/10/2023] [Indexed: 04/19/2023]
Abstract
Matrix metalloproteinases (MMPs) are key drivers of various diseases, including cancer. Development of probes and drugs capable of selectively inhibiting the individual members of the large MMP family remains a persistent challenge. The inhibitory N-terminal domain of tissue inhibitor of metalloproteinases-2 (N-TIMP2), a natural broad MMP inhibitor, can provide a scaffold for protein engineering to create more selective MMP inhibitors. Here, we pursued a unique approach harnessing both computational design and combinatorial screening to confer high binding specificity toward a target MMP in preference to an anti-target MMP. We designed a loop extension of N-TIMP2 to allow new interactions with the non-conserved MMP surface and generated an efficient focused library for yeast surface display, which was then screened for high binding to the target MMP-14 and low binding to anti-target MMP-3. Deep sequencing analysis identified the most promising variants, which were expressed, purified, and tested for selectivity of inhibition. Our best N-TIMP2 variant exhibited 29 pM binding affinity to MMP-14 and 2.4 µM affinity to MMP-3, revealing 7500-fold greater specificity than WT N-TIMP2. High-confidence structural models were obtained by including NGS data in the AlphaFold multiple sequence alignment. The modeling together with experimental mutagenesis validated our design predictions, demonstrating that the loop extension packs tightly against non-conserved residues on MMP-14 and clashes with MMP-3. This study demonstrates how introduction of loop extensions in a manner guided by target protein conservation data and loop design can offer an attractive strategy to achieve specificity in design of protein ligands.
Collapse
Affiliation(s)
- Alessandro Bonadio
- Department of Biological Chemistry, The Alexander Silberman Institute of Life Sciences, The Hebrew University of Jerusalem, Israel
| | - Bernhard L Wenig
- Department of Cancer Biology, Mayo Clinic Comprehensive Cancer Center, Jacksonville, Florida, USA; Paracelsus Medical University, Salzburg, Austria
| | - Alexandra Hockla
- Department of Cancer Biology, Mayo Clinic Comprehensive Cancer Center, Jacksonville, Florida, USA
| | - Evette S Radisky
- Department of Cancer Biology, Mayo Clinic Comprehensive Cancer Center, Jacksonville, Florida, USA.
| | - Julia M Shifman
- Department of Biological Chemistry, The Alexander Silberman Institute of Life Sciences, The Hebrew University of Jerusalem, Israel.
| |
Collapse
|
12
|
Rezhdo A, Lessard CT, Islam M, Van Deventer JA. Strategies for enriching and characterizing proteins with inhibitory properties on the yeast surface. Protein Eng Des Sel 2023; 36:gzac017. [PMID: 36648434 PMCID: PMC10365883 DOI: 10.1093/protein/gzac017] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2022] [Revised: 10/20/2022] [Accepted: 11/07/2022] [Indexed: 01/18/2023] Open
Abstract
Display technologies are powerful tools for discovering binding proteins against a broad range of biological targets. However, it remains challenging to adapt display technologies for the discovery of proteins that inhibit the enzymatic activities of targets. Here, we investigate approaches for discovering and characterizing inhibitory antibodies in yeast display format using a well-defined series of constructs and the target matrix metalloproteinase-9. Three previously reported antibodies were used to create model libraries consisting of inhibitory, non-inhibitory, and non-binding constructs. Conditions that preferentially enrich for inhibitory clones were identified for both magnetic bead-based enrichments and fluorescence-activated cell sorting. Half maximal inhibitory concentration (IC50) was obtained through yeast titration assays. The IC50 of the inhibitory antibody obtained in yeast display format falls within the confidence interval of the IC50 value determined in soluble form. Overall, this study identifies strategies for the discovery and characterization of inhibitory clones directly in yeast display format.
Collapse
Affiliation(s)
- Arlinda Rezhdo
- Chemical and Biological Engineering Department, Tufts University, Medford, MA 02155, USA
| | - Catherine T Lessard
- Chemical and Biological Engineering Department, Tufts University, Medford, MA 02155, USA
| | - Mariha Islam
- Chemical and Biological Engineering Department, Tufts University, Medford, MA 02155, USA
| | - James A Van Deventer
- Chemical and Biological Engineering Department, Tufts University, Medford, MA 02155, USA
- Biomedical Engineering Department, Tufts University, Medford, MA 02155, USA
| |
Collapse
|
13
|
Kwon MJ. Matrix metalloproteinases as therapeutic targets in breast cancer. Front Oncol 2023; 12:1108695. [PMID: 36741729 PMCID: PMC9897057 DOI: 10.3389/fonc.2022.1108695] [Citation(s) in RCA: 18] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2022] [Accepted: 12/28/2022] [Indexed: 01/22/2023] Open
Abstract
Matrix metalloproteinases (MMPs) are the most prominent proteinases involved in tumorigenesis. They were initially recognized to promote tumor progression by remodeling the extracellular matrix through their proteolytic activity. However, accumulating evidence has revealed that some MMPs have protective roles in cancer progression, and the same MMP can exert opposing roles depending on the cell type in which it is expressed or the stage of cancer. Moreover, studies have shown that MMPs are involved in cancer progression through their roles in other biological processes such as cell signaling and immune regulation, independent of their catalytic activity. Despite the prognostic significance of tumoral or stromal expression of MMPs in breast cancer, their roles and molecular mechanisms in breast cancer progression remain unclear. As the failures of early clinical trials with broad-spectrum MMP inhibitors were mainly due to a lack of drug specificity, substantial efforts have been made to develop highly selective MMP inhibitors. Some recently developed MMP inhibitory monoclonal antibodies demonstrated promising anti-tumor effects in preclinical models of breast cancer. Importantly, anti-tumor effects of these antibodies were associated with the modulation of tumor immune microenvironment, suggesting that the use of MMP inhibitors in combination with immunotherapy can improve the efficacy of immunotherapy in HER2-positive or triple-negative breast cancer. In this review, the current understanding of the roles of tumoral or stromal MMPs in breast cancer is summarized, and recent advances in the development of highly selective MMP inhibitors are discussed.
Collapse
Affiliation(s)
- Mi Jeong Kwon
- Vessel-Organ Interaction Research Center (MRC), College of Pharmacy, Kyungpook National University, Daegu, Republic of Korea,BK21 FOUR Community-Based Intelligent Novel Drug Discovery Education Unit, College of Pharmacy and Research Institute of Pharmaceutical Sciences, Kyungpook National University, Daegu, Republic of Korea,*Correspondence: Mi Jeong Kwon,
| |
Collapse
|
14
|
Kim MH, Lim SH. Matrix Metalloproteinases and Glaucoma. Biomolecules 2022; 12:biom12101368. [PMID: 36291577 PMCID: PMC9599265 DOI: 10.3390/biom12101368] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2022] [Revised: 09/20/2022] [Accepted: 09/22/2022] [Indexed: 11/19/2022] Open
Abstract
Matrix metalloproteinases (MMPs) are enzymes that decompose extracellular matrix (ECM) proteins. MMPs are thought to play important roles in cellular processes, such as cell proliferation, differentiation, angiogenesis, migration, apoptosis, and host defense. MMPs are distributed in almost all intraocular tissues and are involved in physiological and pathological mechanisms of the eye. MMPs are also associated with glaucoma, a progressive neurodegenerative disease of the eyes. MMP activity affects intraocular pressure control and apoptosis of retinal ganglion cells, which are the pathological mechanisms of glaucoma. It also affects the risk of glaucoma development based on genetic pleomorphism. In addition, MMPs may affect the treatment outcomes of glaucoma, including the success rate of surgical treatment and side effects on the ocular surface due to glaucoma medications. This review discusses the various relationships between MMP and glaucoma.
Collapse
Affiliation(s)
- Moo Hyun Kim
- Department of Ophthalmology, Daegu Premier Eye Center, Suseong-ro 197, Suseong-Gu, Daegu 42153, Korea
| | - Su-Ho Lim
- Department of Ophthalmology, Daegu Veterans Health Service Medical Center, 60 Wolgok-Ro, Dalseo-Gu, Daegu 42835, Korea
- Correspondence: ; Tel.: +82-53-630-7572
| |
Collapse
|
15
|
Aschner Y, Downey GP. Proteinases in the pathogenesis of lymphangioleiomyomatosis lung disease: nibbling or chewing up the lung? Eur Respir J 2022; 59:2200405. [PMID: 35422429 DOI: 10.1183/13993003.00405-2022] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2022] [Accepted: 03/17/2022] [Indexed: 11/05/2022]
Affiliation(s)
- Yael Aschner
- Division of Pulmonary Sciences and Critical Care Medicine, Department of Medicine, University of Colorado Anschutz Medical Campus, Aurora, CO, USA
| | - Gregory P Downey
- Division of Pulmonary Sciences and Critical Care Medicine, Department of Medicine, University of Colorado Anschutz Medical Campus, Aurora, CO, USA
- Department of Immunology and Microbiology, University of Colorado Anschutz Medical Campus, Aurora, CO, USA
- Departments of Medicine, Pediatrics and Immunology and Genomic Medicine, National Jewish Health, Denver, CO, USA
| |
Collapse
|