1
|
Cazzaniga G, Mori M, Griego A, Scarpa E, Moschetti G, Muzzioli S, Stelitano G, Chiarelli LR, Cocorullo M, Casali E, Porta A, Zanoni G, Tresoldi A, Pini E, Batalha ÍL, Battaglia G, Tuccinardi T, Rizzello L, Villa S, Meneghetti F. Nanoenabling MbtI Inhibitors for Next-Generation Tuberculosis Therapy. J Med Chem 2025; 68:5312-5332. [PMID: 40029993 PMCID: PMC11912484 DOI: 10.1021/acs.jmedchem.4c02386] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2024] [Revised: 02/17/2025] [Accepted: 02/20/2025] [Indexed: 03/14/2025]
Abstract
The urgent need for safer and innovative antitubercular agents remains a priority for the scientific community. In pursuit of this goal, we designed and evaluated novel 5-phenylfuran-2-carboxylic acid derivatives targeting Mycobacterium tuberculosis (Mtb) salicylate synthase (MbtI), a key enzyme, absent in humans, that plays a crucial role in Mtb virulence. Several potent MbtI inhibitors demonstrating significant antitubercular activity and a favorable safety profile were identified. Structure-guided optimization yielded 5-(3-cyano-5-isobutoxyphenyl)furan-2-carboxylic acid (1e), which exhibited strong MbtI inhibition (IC50 = 11.2 μM) and a promising in vitro antitubercular activity (MIC99 = 32 μM against M. bovis BCG). Esters of 1e were effectively loaded into poly(2-methacryloyloxyethyl phosphorylcholine)-poly(2-(diisopropylamino)ethyl methacrylate) (PMPC-PDPA) polymersomes (POs) and delivered to intracellular mycobacteria, resulting in reduced Mtb viability. This study provides a foundation for the use of POs in the development of future MbtI-targeted therapies for tuberculosis.
Collapse
Affiliation(s)
- Giulia Cazzaniga
- Department of Pharmaceutical Sciences, University of Milan, Via L. Mangiagalli 25, 20133 Milano, Italy
- Department of Science and High Technology, University of Insubria, via Valleggio 9, 22100 Como, Italy
| | - Matteo Mori
- Department of Pharmaceutical Sciences, University of Milan, Via L. Mangiagalli 25, 20133 Milano, Italy
| | - Anna Griego
- Department of Pharmaceutical Sciences, University of Milan, Via L. Mangiagalli 25, 20133 Milano, Italy
- National Institute of Molecular Genetic (INGM), Via F. Sforza 35, 20122 Milano, Italy
| | - Edoardo Scarpa
- Department of Pharmaceutical Sciences, University of Milan, Via L. Mangiagalli 25, 20133 Milano, Italy
- National Institute of Molecular Genetic (INGM), Via F. Sforza 35, 20122 Milano, Italy
| | - Giorgia Moschetti
- Department of Pharmaceutical Sciences, University of Milan, Via L. Mangiagalli 25, 20133 Milano, Italy
- National Institute of Molecular Genetic (INGM), Via F. Sforza 35, 20122 Milano, Italy
| | - Stefano Muzzioli
- Department of Pharmaceutical Sciences, University of Milan, Via L. Mangiagalli 25, 20133 Milano, Italy
- National Institute of Molecular Genetic (INGM), Via F. Sforza 35, 20122 Milano, Italy
| | - Giovanni Stelitano
- Department of Biology and Biotechnology "Lazzaro Spallanzani″, University of Pavia, via A. Ferrata 9, 27100 Pavia, Italy
| | - Laurent R Chiarelli
- Department of Biology and Biotechnology "Lazzaro Spallanzani″, University of Pavia, via A. Ferrata 9, 27100 Pavia, Italy
| | - Mario Cocorullo
- Department of Biology and Biotechnology "Lazzaro Spallanzani″, University of Pavia, via A. Ferrata 9, 27100 Pavia, Italy
| | - Emanuele Casali
- Department of Chemistry, University of Pavia, Viale T. Taramelli 12, 27100 Pavia, Italy
| | - Alessio Porta
- Department of Chemistry, University of Pavia, Viale T. Taramelli 12, 27100 Pavia, Italy
| | - Giuseppe Zanoni
- Department of Chemistry, University of Pavia, Viale T. Taramelli 12, 27100 Pavia, Italy
| | - Andrea Tresoldi
- Department of Pharmaceutical Sciences, University of Milan, Via L. Mangiagalli 25, 20133 Milano, Italy
| | - Elena Pini
- Department of Pharmaceutical Sciences, University of Milan, Via L. Mangiagalli 25, 20133 Milano, Italy
| | - Íris L Batalha
- Department of Life Sciences, University of Bath, Claverton Down, BA2 7AY Bath, U.K
| | - Giuseppe Battaglia
- Molecular Bionics Group, Institute for Bioengineering of Catalonia (IBEC), C. Baldiri Reixac 10-12, 08028 Barcelona, Spain
- Catalan Institution of Research and Advanced Studies, (ICREA), Passeig de Lluís Companys, 23, 08010 Barcelona, Spain
| | - Tiziano Tuccinardi
- Department of Pharmacy, University of Pisa, Via Bonanno Pisano 6, 56126 Pisa, Italy
| | - Loris Rizzello
- Department of Pharmaceutical Sciences, University of Milan, Via L. Mangiagalli 25, 20133 Milano, Italy
- National Institute of Molecular Genetic (INGM), Via F. Sforza 35, 20122 Milano, Italy
| | - Stefania Villa
- Department of Pharmaceutical Sciences, University of Milan, Via L. Mangiagalli 25, 20133 Milano, Italy
| | - Fiorella Meneghetti
- Department of Pharmaceutical Sciences, University of Milan, Via L. Mangiagalli 25, 20133 Milano, Italy
| |
Collapse
|
2
|
Mokrousov I, Angelova VT, Slavchev I, Bezruchko MV, Dimitrov S, Polev DE, Dobrikov GM, Valcheva V. Genomic Insight into Primary Adaptation of Mycobacterium tuberculosis to Aroylhydrazones and Nitrofuroylamides In Vitro. Antibiotics (Basel) 2025; 14:225. [PMID: 40149037 PMCID: PMC11939388 DOI: 10.3390/antibiotics14030225] [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: 02/01/2025] [Revised: 02/17/2025] [Accepted: 02/20/2025] [Indexed: 03/29/2025] Open
Abstract
Background/Objectives: New anti-tuberculosis compounds are needed to treat patients infected with multi- or extensively drug-resistant Mycobacterium tuberculosis strains. Studies based on spontaneous in vitro mutagenesis can provide insights into the possible modes of action and resistance mechanisms of such new compounds. We evaluated the primary response of M. tuberculosis in vitro to the action of new aroylhydrazones and nitrofuroylamides. Methods: The reference strain H37Rv was cultured on solid media with compounds at increased concentrations relative to MIC. Resistant clones were investigated using whole-genome sequencing and bioinformatics tools to assess the role and potential impact of identified mutations. Results: Some of the mutations are significant (based on in silico analysis), located in essential genes, and therefore of particular interest. Frameshift mutations were observed in (i) Rv2702/ppgK, which is associated with starvation-induced drug tolerance and persistence in mice, and (ii) Rv3696c/glpK, which has been described as a switch on/off mutation associated with drug tolerance. Nonsynonymous substitutions were found in Rv0506/mmpS2, which belongs to the Mmp protein family involved in transport and drug efflux, and in infB, encoding the translation initiation factor IF-2. Conclusions: The primary adaptation of M. tuberculosis to the selective pressure of the tested compounds is complex and multifaceted. It involves multiple unrelated genes and pathways linked to non-specific drug tolerance, efflux systems, or mechanisms counteracting oxidative stress.
Collapse
Affiliation(s)
- Igor Mokrousov
- St. Petersburg Pasteur Institute, 197101 St. Petersburg, Russia; (M.V.B.); (D.E.P.)
| | | | - Ivaylo Slavchev
- Institute of Organic Chemistry with Centre of Phytochemistry, Bulgarian Academy of Sciences, 1113 Sofia, Bulgaria; (I.S.); (G.M.D.)
| | - Mikhail V. Bezruchko
- St. Petersburg Pasteur Institute, 197101 St. Petersburg, Russia; (M.V.B.); (D.E.P.)
| | - Simeon Dimitrov
- The Stephan Angeloff Institute of Microbiology, Bulgarian Academy of Sciences, 1113 Sofia, Bulgaria;
| | - Dmitrii E. Polev
- St. Petersburg Pasteur Institute, 197101 St. Petersburg, Russia; (M.V.B.); (D.E.P.)
| | - Georgi M. Dobrikov
- Institute of Organic Chemistry with Centre of Phytochemistry, Bulgarian Academy of Sciences, 1113 Sofia, Bulgaria; (I.S.); (G.M.D.)
| | - Violeta Valcheva
- The Stephan Angeloff Institute of Microbiology, Bulgarian Academy of Sciences, 1113 Sofia, Bulgaria;
| |
Collapse
|
3
|
Terreiro JFPR, Marquês JT, Antunes I, de Faria CF, Santos S, Martins F, de Almeida RFM. Membrane interaction studies of isoniazid derivatives active against drug-resistant tuberculosis. Eur J Pharm Sci 2025; 205:106986. [PMID: 39674553 DOI: 10.1016/j.ejps.2024.106986] [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: 09/19/2024] [Revised: 11/22/2024] [Accepted: 12/09/2024] [Indexed: 12/16/2024]
Abstract
Tuberculosis is one of the leading causes of mortality worldwide due to the growth of multi-drug resistant strains unsusceptible to currently available therapies. Four compounds, isoniazid (INH) and three derivatives, N'-decanoylisonicotinohydrazide (INHC10), N'-(E)-(4-phenoxybenzylidene)isonicotinohydrazide (N34) and N'-(4-phenoxybenzyl)isonicotinohydrazide (N34red), were studied. Owing to their advantageous in vitro selectivity index against the primary mutation responsible for drug resistance in Mycobacterium tuberculosis (Mtb), as well as their suitable lipophilicity and interaction with human serum albumin, INHC10 and N34 were deemed promising antitubercular compounds. N34red, despite differing from N34 only in the saturation of the N' = C bond, presents a poor selectivity index. To delve deeper into the therapeutic potential of these compounds, their interaction with biomembrane models, mimicking biological barriers on the way to the target inside Mtb cells, was herein evaluated. All compounds, except N34red, weakened the packing of the acyl chains in the rigid lipid gel phase, especially INHC10, which was the only compound disturbing liquid disordered membranes. Notably, all compounds except INH decreased membrane dipole potential, across all types of bilayers studied, but only N34red had a drastic effect. The insertion in gel phase bilayers suggests that the compounds may be able to penetrate the rigid cell wall of Mtb. Förster's resonance energy transfer (FRET) assays in ternary bilayers with liquid ordered/liquid disordered lateral heterogeneity mimicking human cell membranes, showed that the compounds affected neither the size nor the organization of lipid domains. These results provide molecular insights into the low toxicity against human cell lines and improved activity against drug-resistant Mtb of INHC10 and N34.
Collapse
Affiliation(s)
- Joana F P R Terreiro
- Departamento de Química e Bioquímica, Faculdade de Ciências, Centro de Química Estrutural, Institute of Molecular Sciences, Universidade de Lisboa, Campo Grande 1749-016, Portugal
| | - Joaquim T Marquês
- Departamento de Química e Bioquímica, Faculdade de Ciências, Centro de Química Estrutural, Institute of Molecular Sciences, Universidade de Lisboa, Campo Grande 1749-016, Portugal.
| | - Inês Antunes
- Departamento de Química e Bioquímica, Faculdade de Ciências, Centro de Química Estrutural, Institute of Molecular Sciences, Universidade de Lisboa, Campo Grande 1749-016, Portugal
| | - Catarina Frazão de Faria
- Departamento de Química e Bioquímica, Faculdade de Ciências, Centro de Química Estrutural, Institute of Molecular Sciences, Universidade de Lisboa, Campo Grande 1749-016, Portugal
| | - Susana Santos
- Departamento de Química e Bioquímica, Faculdade de Ciências, Centro de Química Estrutural, Institute of Molecular Sciences, Universidade de Lisboa, Campo Grande 1749-016, Portugal
| | - Filomena Martins
- Departamento de Química e Bioquímica, Faculdade de Ciências, Centro de Química Estrutural, Institute of Molecular Sciences, Universidade de Lisboa, Campo Grande 1749-016, Portugal.
| | - Rodrigo F M de Almeida
- Departamento de Química e Bioquímica, Faculdade de Ciências, Centro de Química Estrutural, Institute of Molecular Sciences, Universidade de Lisboa, Campo Grande 1749-016, Portugal.
| |
Collapse
|
4
|
Shirazi SHUA, Rizvi F, Sherwani ZA, Siddiqui AR, Ul-Haq Z, Siddiqui H, Choudhary MI, Ahmad MS. Synthesis of isoniazid derivatives and evaluation of their α-chymotrypsin inhibitory effect through in silico guided in vitro studies. Biochem Biophys Res Commun 2024; 735:150805. [PMID: 39426135 DOI: 10.1016/j.bbrc.2024.150805] [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: 07/10/2024] [Revised: 10/08/2024] [Accepted: 10/08/2024] [Indexed: 10/21/2024]
Abstract
Alpha-chymotrypsin is a serine protease. Its overexpression is responsible for several ailments, such as chronic obstructive pulmonary disease, autoimmune diseases, pancreatitis, and colon cancers. Therefore, the discovery of potent α-chymotrypsin inhibitors is essential for the treatment of the aforementioned ailments. In this study, we identified new α-chymotrypsin inhibitors through a systematic approach, utilizing the in silico and in vivo studies to predict and confirm the inhibitory potential of isoniazid derivatives. During this study, six compounds 2, 3, 4, 7, 9, and 10 were shortlisted from ten isoniazid derivatives through in silico screening. After that, MD simulations were performed for these compounds. The shortlisted compounds were evaluated through an in vitro α-chymotrypsin inhibitory assay. Compounds 9 and 10 showed a potent inhibition against α-chymotrypsin. The identified compounds or their derivatives can be further investigated as drug leads against the ailments caused by α-chymotrypsin and related serine proteases.
Collapse
Affiliation(s)
- Syeda Hoor-Ul-Ain Shirazi
- H. E. J. Research Institute of Chemistry, International Center for Chemical and Biological Sciences, University of Karachi, Karachi, 75270, Pakistan
| | - Fazila Rizvi
- H. E. J. Research Institute of Chemistry, International Center for Chemical and Biological Sciences, University of Karachi, Karachi, 75270, Pakistan
| | - Zaid Anis Sherwani
- Dr. Panjwani Center for Molecular Medicine and Drug Research, International Center for Chemical and Biological Sciences, University of Karachi, Karachi, 75270, Pakistan
| | - Ali Raza Siddiqui
- H. E. J. Research Institute of Chemistry, International Center for Chemical and Biological Sciences, University of Karachi, Karachi, 75270, Pakistan
| | - Zaheer Ul-Haq
- Dr. Panjwani Center for Molecular Medicine and Drug Research, International Center for Chemical and Biological Sciences, University of Karachi, Karachi, 75270, Pakistan
| | - Hina Siddiqui
- H. E. J. Research Institute of Chemistry, International Center for Chemical and Biological Sciences, University of Karachi, Karachi, 75270, Pakistan
| | - M Iqbal Choudhary
- H. E. J. Research Institute of Chemistry, International Center for Chemical and Biological Sciences, University of Karachi, Karachi, 75270, Pakistan; Dr. Panjwani Center for Molecular Medicine and Drug Research, International Center for Chemical and Biological Sciences, University of Karachi, Karachi, 75270, Pakistan
| | - Malik Shoaib Ahmad
- Dr. Panjwani Center for Molecular Medicine and Drug Research, International Center for Chemical and Biological Sciences, University of Karachi, Karachi, 75270, Pakistan.
| |
Collapse
|
5
|
Rouzi K, Altay A, Bouatia M, Yeniçeri E, Islam MS, Oulmidi A, El Karbane M, Karrouchi K. Novel isoniazid-hydrazone derivatives induce cell growth inhibition, cell cycle arrest and apoptosis via mitochondria-dependent caspase activation and PI3K/AKT inhibition. Bioorg Chem 2024; 150:107563. [PMID: 38885547 DOI: 10.1016/j.bioorg.2024.107563] [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: 03/25/2024] [Revised: 05/13/2024] [Accepted: 06/13/2024] [Indexed: 06/20/2024]
Abstract
In this study, seven isoniazid-hydrazone derivatives (3a-g) were synthesized and their structures elucidated by chromatographic techniques, and then the antiproliferative effects of these compounds on various cancer cells were tested. The advanced anticancer mechanism of the most potent compound was then investigated. Antiproliferative activities of the synthesized compounds were evaluated on human breast cancer MCF-7, lung cancer A-549, colon cancer HT-29, and non-cancerous mouse fibroblast 3T3-L1 cell lines by XTT assay. Flow cytometry analysis were carried out to determine cell cycle distribution, apoptosis, mitochondrial membrane potential, multi-caspase activity, and expression of PI3K/AKT signaling pathway. The XTT results showed that all the title molecules displayed cytotoxic activity at varying strengths in different dose ranges, and among them, the strongest cytotoxic effect and high selectivity were exerted by 3d against MCF-7 cells with the IC50 value of 11.35 µM and selectivity index of 8.65. Flow cytometry results revealed that compound 3d induced apoptosis through mitochondrial membrane disruption and multi-caspase activation in MCF-7 cells. It also inhibited the cell proliferation via inhibition of expression of PI3K/AKT and arrested the cell cycle at G0/G1 phase. In conclusion, all these data disclosed that among the synthesized compounds, 3d is notable for in vivo anticancer studies.
Collapse
Affiliation(s)
- Khouloud Rouzi
- Laboratory of Analytical Chemistry and Bromatology, Team of Formulation and Quality Control of Health Products, Faculty of Medicine and Pharmacy, Mohammed V University in Rabat, Morocco
| | - Ahmet Altay
- Department of Chemistry, Faculty of Arts and Science, Erzincan Binali Yıldırım University, 24100 Erzincan, Turkey.
| | - Mustapha Bouatia
- Laboratory of Analytical Chemistry and Bromatology, Team of Formulation and Quality Control of Health Products, Faculty of Medicine and Pharmacy, Mohammed V University in Rabat, Morocco
| | - Esma Yeniçeri
- Department of Chemistry, Institute of Science and Technology, Erzincan Binali Yıldırım University, 24002 Erzincan, Turkey
| | - Mohammad Shahidul Islam
- College of Science, Chemistry Department, King Saud University, P.O. Box 2455, Riyadh 11451, Saudi Arabia
| | - Afaf Oulmidi
- Institute of Condensed Matter and Nanosciences, Molecular Chemistry, Materials and Catalysis (IMCN/MOST), Université Catholique de Louvain, 1348 Louvain-la-Neuve, Belgium
| | - Miloud El Karbane
- Laboratory of Analytical Chemistry and Bromatology, Team of Formulation and Quality Control of Health Products, Faculty of Medicine and Pharmacy, Mohammed V University in Rabat, Morocco
| | - Khalid Karrouchi
- Laboratory of Analytical Chemistry and Bromatology, Team of Formulation and Quality Control of Health Products, Faculty of Medicine and Pharmacy, Mohammed V University in Rabat, Morocco.
| |
Collapse
|
6
|
Basu S, Mandal S, Maiti PK. Permeability of TB drugs through the mycolic acid monolayer: a tale of two force fields. Phys Chem Chem Phys 2024; 26:21429-21440. [PMID: 39101468 DOI: 10.1039/d4cp02659d] [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: 08/06/2024]
Abstract
Tuberculosis (TB) treatment becomes challenging due to the unique cell wall structure of Mycobacterium tuberculosis (M. tb). Among various components of the M.tb cell wall, mycolic acid (MA) is of particular interest because it is speculated to exhibit extremely low permeability for most of the drug molecules, thus helping M.tb to survive against medical treatment. However, no quantitative assessment of the thermodynamic barrier encountered by various well-known TB drugs in the mycolic acid monolayer has been performed so far using computational tools. On this premise, our present work aims to probe the permeability of some first and second line TB drugs, namely ethambutol, ethionamide, and isoniazid, through the modelled mycolic acid monolayer, using molecular dynamics (MD) simulation with two sets of force field (FF) parameters, namely GROMOS 54A7-ATB (GROMOS) and CHARMM36 (CHARMM) FFs. Our findings indicate that both FFs provide consistent results in terms of the mode of drug-monolayer interactions but significantly differ in the drug permeability through the monolayer. The mycolic acid monolayer generally exhibited a higher free energy barrier of crossing with CHARMM FF, while with GROMOS FF, better stability of drug molecules on the monolayer surface was observed, which can be attributed to the greater electrostatic potential at the monolayer-water interface, found for the later. Although both the FF parameters predicted the highest resistance against ethambutol (permeability values of 8.40 × 10-34 cm s-1 and 9.61 × 10-31 cm s-1 for the CHARMM FF and the GROMOS FF, respectively), results obtained using GROMOS were found to be consistent with the water solubility of drugs, suggesting it to be a slightly better FF for modelling drug-mycolic acid interactions. Therefore, this study enhances our understanding of TB drug permeability and highlights the potential of the GROMOS FF in simulating drug-mycolic acid interactions.
Collapse
Affiliation(s)
- Subhadip Basu
- Centre for Condensed Matter Theory, Department of Physics, Indian Institute of Science, Bangalore, India.
| | - Sandip Mandal
- Centre for Condensed Matter Theory, Department of Physics, Indian Institute of Science, Bangalore, India.
| | - Prabal K Maiti
- Centre for Condensed Matter Theory, Department of Physics, Indian Institute of Science, Bangalore, India.
| |
Collapse
|
7
|
Gavadia R, Rasgania J, Sahu N, Varma-Basil M, Chauhan V, Kumar S, Mor S, Singh D, Jakhar K. Synthesis of indole-functionalized isoniazid conjugates with potent antimycobacterial and antioxidant efficacy. Future Med Chem 2024; 16:1731-1747. [PMID: 39041719 PMCID: PMC11457681 DOI: 10.1080/17568919.2024.2379240] [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: 03/05/2024] [Accepted: 06/14/2024] [Indexed: 07/24/2024] Open
Abstract
Aim: Developing potent medicinal alternates for tuberculosis (TB) is highly desirable due to the advent of drug-resistant lethal TB strains.Methods & results: Novel indole-isoniazid integrates have been synthesized with promising antimycobacterial action against the H37Rv strain, and the nitro analogs 4e and 4j show the highest efficacy with a minimum inhibitory concentration of 1.25 μg/ml. The molecular docking studies against InhA support the experimental findings. Indole conjugates display remarkable radical quenching efficiency, and compounds 4e and 4j demonstrate maximum IC50 values of 50.19 and 52.45 μg/ml, respectively. Pharmacokinetic analysis anticipated appreciable druggability for the title compounds.Conclusion: The notable bioaction of the indole-isoniazid templates projects them as potential lead in developing anti-TB medications with synergetic antioxidant action.
Collapse
Affiliation(s)
- Renu Gavadia
- Department of Chemistry, M. D. University, Rohtak, Haryana, 124001, India
| | - Jyoti Rasgania
- Department of Chemistry, M. D. University, Rohtak, Haryana, 124001, India
| | - Neetu Sahu
- Department of Chemistry, M. D. University, Rohtak, Haryana, 124001, India
| | - Mandira Varma-Basil
- Department of Microbiology, Vallabhbhai Patel Chest Institute, University of Delhi, Delhi, 110007, India
| | - Varsha Chauhan
- Department of Microbiology, Vallabhbhai Patel Chest Institute, University of Delhi, Delhi, 110007, India
- Department of Microbiology, M. D. UniversityRohtak, Haryana, 124001, India
| | - Sanjay Kumar
- Department of Microbiology, M. D. UniversityRohtak, Haryana, 124001, India
| | - Satbir Mor
- Department of Chemistry, Guru Jambheshwar University of Science & Technology, Hisar, Haryana, 125001, India
| | - Devender Singh
- Department of Chemistry, M. D. University, Rohtak, Haryana, 124001, India
| | - Komal Jakhar
- Department of Chemistry, M. D. University, Rohtak, Haryana, 124001, India
| |
Collapse
|
8
|
Franco Machado J, Sá M, Pires I, da Silva MT, Marques F, Coelho JAS, Mendes F, Piedade MFM, Machuqueiro M, Jiménez MA, Garcia MH, Correia JDG, Morais TS. Dual FGFR-targeting and pH-activatable ruthenium-peptide conjugates for targeted therapy of breast cancer. Dalton Trans 2024; 53:7682-7693. [PMID: 38573236 DOI: 10.1039/d4dt00497c] [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: 04/05/2024]
Abstract
Dysregulation of Fibroblast Growth Factor Receptors (FGFRs) signaling has been associated with breast cancer, yet employing FGFR-targeted delivery systems to improve the efficacy of cytotoxic agents is still sparsely exploited. Herein, we report four new bi-functional ruthenium-peptide conjugates (RuPCs) with FGFR-targeting and pH-dependent releasing abilities, envisioning the selective delivery of cytotoxic Ru complexes to FGFR(+)-breast cancer cells, and controlled activation at the acidic tumoral microenvironment. The antiproliferative potential of the RuPCs and free Ru complexes was evaluated in four breast cancer cell lines with different FGFR expression levels (SKBR-3, MDA-MB-134-VI, MCF-7, and MDA-MB-231) and in human dermal fibroblasts (HDF), at pH 6.8 and pH 7.4 aimed at mimicking the tumor microenvironment and normal tissues/bloodstream pHs, respectively. The RuPCs showed higher cytotoxicity in cells with higher level of FGFR expression at acidic pH. Additionally, RuPCs showed up to 6-fold higher activity in the FGFR(+) breast cancer lines compared to the normal cell line. The release profile of Ru complexes from RuPCs corroborates the antiproliferative effects observed. Remarkably, the cytotoxicity and releasing ability of RuPCs were shown to be strongly dependent on the conjugation of the peptide position in the Ru complex. Complementary molecular dynamic simulations and computational calculations were performed to help interpret these findings at the molecular level. In summary, we identified a lead bi-functional RuPC that holds strong potential as a FGFR-targeted chemotherapeutic agent.
Collapse
Affiliation(s)
- João Franco Machado
- Centro de Química Estrutural, Institute of Molecular Sciences, Faculdade de Ciências, Universidade de Lisboa, Campo Grande, 1749-016 Lisboa, Portugal.
- Centro de Ciências e Tecnologias Nucleares, Instituto Superior Técnico, Universidade de Lisboa, Estrada Nacional 10 (km 139, 7), 2695-066 Bobadela LRS, Portugal.
| | - Marco Sá
- Centro de Química Estrutural, Institute of Molecular Sciences, Faculdade de Ciências, Universidade de Lisboa, Campo Grande, 1749-016 Lisboa, Portugal.
| | - Inês Pires
- BioISI - Biosystems & Integrative Sciences Institute, Faculdade de Ciências, Universidade de Lisboa, Campo Grande, 1749-016 Lisboa, Portugal
| | - Miguel Tarita da Silva
- Centro de Ciências e Tecnologias Nucleares, Instituto Superior Técnico, Universidade de Lisboa, Estrada Nacional 10 (km 139, 7), 2695-066 Bobadela LRS, Portugal.
| | - Fernanda Marques
- Centro de Ciências e Tecnologias Nucleares, Instituto Superior Técnico, Universidade de Lisboa, Estrada Nacional 10 (km 139, 7), 2695-066 Bobadela LRS, Portugal.
- Departamento de Engenharia e Ciências Nucleares, Instituto Superior Técnico, Universidade de Lisboa, Estrada Nacional 10 (km 139, 7), 2695-066 Bobadela LRS, Portugal
| | - Jaime A S Coelho
- Centro de Química Estrutural, Institute of Molecular Sciences, Faculdade de Ciências, Universidade de Lisboa, Campo Grande, 1749-016 Lisboa, Portugal.
| | - Filipa Mendes
- Centro de Ciências e Tecnologias Nucleares, Instituto Superior Técnico, Universidade de Lisboa, Estrada Nacional 10 (km 139, 7), 2695-066 Bobadela LRS, Portugal.
- Departamento de Engenharia e Ciências Nucleares, Instituto Superior Técnico, Universidade de Lisboa, Estrada Nacional 10 (km 139, 7), 2695-066 Bobadela LRS, Portugal
| | - M Fátima M Piedade
- Departamento de Química e Bioquímica, Faculdade de Ciências, Universidade de Lisboa, Campo Grande, 1749-016 Lisboa, Portugal
- Centro de Química Estrutural, Institute of Molecular Sciences, Instituto Superior Técnico, Universidade de Lisboa, Av. Rovisco Pais, 1049-001 Lisboa, Portugal
| | - Miguel Machuqueiro
- BioISI - Biosystems & Integrative Sciences Institute, Faculdade de Ciências, Universidade de Lisboa, Campo Grande, 1749-016 Lisboa, Portugal
- Departamento de Química e Bioquímica, Faculdade de Ciências, Universidade de Lisboa, Campo Grande, 1749-016 Lisboa, Portugal
| | - María Angeles Jiménez
- Institute of Physical Chemistry Blas Cabreras (IQF-CSIC), Serrano 119, E-28006 Madrid, Spain
| | - Maria Helena Garcia
- Centro de Química Estrutural, Institute of Molecular Sciences, Faculdade de Ciências, Universidade de Lisboa, Campo Grande, 1749-016 Lisboa, Portugal.
- Departamento de Química e Bioquímica, Faculdade de Ciências, Universidade de Lisboa, Campo Grande, 1749-016 Lisboa, Portugal
| | - João D G Correia
- Centro de Ciências e Tecnologias Nucleares, Instituto Superior Técnico, Universidade de Lisboa, Estrada Nacional 10 (km 139, 7), 2695-066 Bobadela LRS, Portugal.
- Departamento de Engenharia e Ciências Nucleares, Instituto Superior Técnico, Universidade de Lisboa, Estrada Nacional 10 (km 139, 7), 2695-066 Bobadela LRS, Portugal
| | - Tânia S Morais
- Centro de Química Estrutural, Institute of Molecular Sciences, Faculdade de Ciências, Universidade de Lisboa, Campo Grande, 1749-016 Lisboa, Portugal.
- Departamento de Química e Bioquímica, Faculdade de Ciências, Universidade de Lisboa, Campo Grande, 1749-016 Lisboa, Portugal
| |
Collapse
|
9
|
Balandis B, Kavaliauskas P, Grybaitė B, Petraitis V, Petraitienė R, Naing E, Garcia A, Grigalevičiūtė R, Mickevičius V. Synthesis of Novel Benzenesulfonamide-Bearing Functionalized Imidazole Derivatives as Novel Candidates Targeting Multidrug-Resistant Mycobacterium abscessus Complex. Microorganisms 2023; 11:microorganisms11040935. [PMID: 37110358 PMCID: PMC10145568 DOI: 10.3390/microorganisms11040935] [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: 03/04/2023] [Revised: 03/27/2023] [Accepted: 04/01/2023] [Indexed: 04/29/2023] Open
Abstract
Infections caused by drug-resistant (DR) Mycobacterium abscessus (M. abscessus) complex (MAC) are an important public health concern, particularly when affecting individuals with various immunodeficiencies or chronic pulmonary diseases. Rapidly growing antimicrobial resistance among MAC urges us to develop novel antimicrobial candidates for future optimization. Therefore, we have designed and synthesized benzenesulfonamide-bearing functionalized imidazole or S-alkylated derivatives and evaluated their antimicrobial activity using multidrug-resistant M. abscessus strains and compared their antimycobacterial activity using M. bovis BCG and M. tuberculosis H37Ra. Benzenesulfonamide-bearing imidazole-2-thiol compound 13, containing 4-CF3 substituent in benzene ring, showed strong antimicrobial activity against the tested mycobacterial strains and was more active than some antibiotics used as a reference. Furthermore, an imidazole-bearing 4-F substituent and S-methyl group demonstrated good antimicrobial activity against M. abscessus complex strains, as well as M. bovis BCG and M. tuberculosis H37Ra. In summary, these results demonstrated that novel benzenesulfonamide derivatives, bearing substituted imidazoles, could be further explored as potential candidates for the further hit-to-lead optimization of novel antimycobacterial compounds.
Collapse
Affiliation(s)
- Benas Balandis
- Department of Organic Chemistry, Kaunas University of Technology, Radvilėnų Rd. 19, LT-50254 Kaunas, Lithuania
| | - Povilas Kavaliauskas
- Department of Organic Chemistry, Kaunas University of Technology, Radvilėnų Rd. 19, LT-50254 Kaunas, Lithuania
- Transplantation-Oncology Infectious Diseases Program, Division of Infectious Diseases, Department of Medicine, Weill Cornell Medicine of Cornell University, 1300 York Ave., New York, NY 10065, USA
- Institute for Genome Sciences, School of Medicine, University of Maryland Baltimore, 655 W. Baltimore Street, Baltimore, MD 21201, USA
- Institute of Infectious Diseases and Pathogenic Microbiology, Birštono Str. 38A, LT-59116 Prienai, Lithuania
- Biological Research Center, Lithuanian University of Health Sciences, Tilžės Str. 18/7, LT-47181 Kaunas, Lithuania
| | - Birutė Grybaitė
- Department of Organic Chemistry, Kaunas University of Technology, Radvilėnų Rd. 19, LT-50254 Kaunas, Lithuania
| | - Vidmantas Petraitis
- Transplantation-Oncology Infectious Diseases Program, Division of Infectious Diseases, Department of Medicine, Weill Cornell Medicine of Cornell University, 1300 York Ave., New York, NY 10065, USA
- Institute of Infectious Diseases and Pathogenic Microbiology, Birštono Str. 38A, LT-59116 Prienai, Lithuania
- Biological Research Center, Lithuanian University of Health Sciences, Tilžės Str. 18/7, LT-47181 Kaunas, Lithuania
| | - Rūta Petraitienė
- Transplantation-Oncology Infectious Diseases Program, Division of Infectious Diseases, Department of Medicine, Weill Cornell Medicine of Cornell University, 1300 York Ave., New York, NY 10065, USA
- Institute of Infectious Diseases and Pathogenic Microbiology, Birštono Str. 38A, LT-59116 Prienai, Lithuania
| | - Ethan Naing
- Transplantation-Oncology Infectious Diseases Program, Division of Infectious Diseases, Department of Medicine, Weill Cornell Medicine of Cornell University, 1300 York Ave., New York, NY 10065, USA
| | - Andrew Garcia
- Transplantation-Oncology Infectious Diseases Program, Division of Infectious Diseases, Department of Medicine, Weill Cornell Medicine of Cornell University, 1300 York Ave., New York, NY 10065, USA
| | - Ramunė Grigalevičiūtė
- Biological Research Center, Lithuanian University of Health Sciences, Tilžės Str. 18/7, LT-47181 Kaunas, Lithuania
| | - Vytautas Mickevičius
- Department of Organic Chemistry, Kaunas University of Technology, Radvilėnų Rd. 19, LT-50254 Kaunas, Lithuania
| |
Collapse
|
10
|
Teneva Y, Simeonova R, Valcheva V, Angelova VT. Recent Advances in Anti-Tuberculosis Drug Discovery Based on Hydrazide-Hydrazone and Thiadiazole Derivatives Targeting InhA. Pharmaceuticals (Basel) 2023; 16:ph16040484. [PMID: 37111241 PMCID: PMC10140854 DOI: 10.3390/ph16040484] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2023] [Revised: 03/15/2023] [Accepted: 03/21/2023] [Indexed: 04/29/2023] Open
Abstract
Tuberculosis is an extremely serious problem of global public health. Its incidence is worsened by the presence of multidrug-resistant (MDR) strains of Mycobacterium tuberculosis. More serious forms of drug resistance have been observed in recent years. Therefore, the discovery and/or synthesis of new potent and less toxic anti-tubercular compounds is very critical, especially having in mind the consequences and the delays in treatment caused by the COVID-19 pandemic. Enoyl-acyl carrier protein reductase (InhA) is an important enzyme involved in the biosynthesis of mycolic acid, a major component of the M. tuberculosis cell wall. At the same time, it is a key enzyme in the development of drug resistance, making it an important target for the discovery of new antimycobacterial agents. Many different chemical scaffolds, including hydrazide hydrazones and thiadiazoles, have been evaluated for their InhA inhibitory activity. The aim of this review is to evaluate recently described hydrazide-hydrazone- and thiadiazole-containing derivatives that inhibit InhA activity, resulting in antimycobacterial effects. In addition, a brief review of the mechanisms of action of currently available anti-tuberculosis drugs is provided, including recently approved agents and molecules in clinical trials.
Collapse
Affiliation(s)
- Yoanna Teneva
- Faculty of Pharmacy, Medical University of Sofia, 1000 Sofia, Bulgaria
| | - Rumyana Simeonova
- Faculty of Pharmacy, Medical University of Sofia, 1000 Sofia, Bulgaria
| | - Violeta Valcheva
- The Stephan Angeloff Institute of Microbiology, Bulgarian Academy of Sciences, 1113 Sofia, Bulgaria
| | | |
Collapse
|
11
|
Gavadia R, Rasgania J, Basil MV, Chauhan V, Kumar S, Jakhar K. Synthesis of Isoniazid analogs with Promising Antituberculosis Activity and Bioavailability: Biological Evaluation and Computational Studies. J Mol Struct 2023. [DOI: 10.1016/j.molstruc.2023.135325] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/12/2023]
|
12
|
Magalhães P, Reis PBPS, Vila-Viçosa D, Machuqueiro M, Victor BL. Optimization of an in Silico Protocol Using Probe Permeabilities to Identify Membrane Pan-Assay Interference Compounds. J Chem Inf Model 2022; 62:3034-3042. [PMID: 35697029 PMCID: PMC9770580 DOI: 10.1021/acs.jcim.2c00372] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
Membrane pan-assay interference compounds (PAINS) are a class of molecules that interact nonspecifically with lipid bilayers and alter their physicochemical properties. An early identification of these compounds avoids chasing false leads and the needless waste of time and resources in drug discovery campaigns. In this work, we optimized an in silico protocol on the basis of umbrella sampling (US)/molecular dynamics (MD) simulations to discriminate between compounds with different membrane PAINS behavior. We showed that the method is quite sensitive to membrane thickness fluctuations, which was mitigated by changing the US reference position to the phosphate atoms of the closest interacting monolayer. The computational efficiency was improved further by decreasing the number of umbrellas and adjusting their strength and position in our US scheme. The inhomogeneous solubility-diffusion model (ISDM) used to calculate the membrane permeability coefficients confirmed that resveratrol and curcumin have distinct membrane PAINS characteristics and indicated a misclassification of nothofagin in a previous work. Overall, we have presented here a promising in silico protocol that can be adopted as a future reference method to identify membrane PAINS.
Collapse
Affiliation(s)
- Pedro
R. Magalhães
- BioISI
- Biosystems & Integrative Sciences Institute, Faculty of Sciences, University of Lisboa, Campo Grande, C8 bdg, 1749-016 Lisboa, Portugal
| | - Pedro B. P. S. Reis
- BioISI
- Biosystems & Integrative Sciences Institute, Faculty of Sciences, University of Lisboa, Campo Grande, C8 bdg, 1749-016 Lisboa, Portugal
| | - Diogo Vila-Viçosa
- BioISI
- Biosystems & Integrative Sciences Institute, Faculty of Sciences, University of Lisboa, Campo Grande, C8 bdg, 1749-016 Lisboa, Portugal
| | - Miguel Machuqueiro
- BioISI
- Biosystems & Integrative Sciences Institute, Faculty of Sciences, University of Lisboa, Campo Grande, C8 bdg, 1749-016 Lisboa, Portugal
| | - Bruno L. Victor
- BioISI
- Biosystems & Integrative Sciences Institute, Faculty of Sciences, University of Lisboa, Campo Grande, C8 bdg, 1749-016 Lisboa, Portugal
| |
Collapse
|
13
|
Designing quinoline-isoniazid hybrids as potent anti-tubercular agents inhibiting mycolic acid biosynthesis. Eur J Med Chem 2022; 239:114531. [PMID: 35759907 DOI: 10.1016/j.ejmech.2022.114531] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2022] [Revised: 05/04/2022] [Accepted: 06/10/2022] [Indexed: 11/23/2022]
Abstract
Isoniazid is a cornerstone of modern tuberculosis (TB) therapy and targets the enoyl ACP reductase InhA, a key enzyme in mycolic acid biosynthesis. InhA is still a promising target for the development of new anti-TB drugs. Herein, we report the design, synthesis, and anti-tubercular activity of new isoniazid hybrids. Among these, 1H-1,2,3 triazole-tethered quinoline-isoniazid conjugates 16a to 16g exhibited high activity against Mycobacterium tuberculosis with minimal inhibitory concentrations in the 0.25-0.50 μg/mL range and were bactericidal in vitro. Importantly, these compounds were well tolerated at high doses on mammalian cells, leading to high selectivity indices. The hybrids were dependent on functional KatG production to inhibit mycolic acid biosynthesis. Moreover, overexpression of InhA in M. tuberculosis resulted in high resistance levels to 16a-16g and reduced mycolic acid biosynthesis inhibition, similar to isoniazid. Overall, these findings suggest that the synthesized quinoline-isoniazid hybrids are promising anti-tubercular molecules, which require further pre-clinical evaluation.
Collapse
|
14
|
Scheepers MC, Fernandes MA, Lemmerer A. Chains or rings? Polymorphism of an isoniazid derivative derivatized with diacetone alcohol. RSC Adv 2022; 12:11658-11664. [PMID: 35432943 PMCID: PMC9008515 DOI: 10.1039/d2ra02057b] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2022] [Accepted: 04/03/2022] [Indexed: 11/29/2022] Open
Abstract
Isoniazid was derivated with diacetone alcohol in a Schiff-base reaction in order to yield N'-[(2E)-4-hydroxy-4-methylpentan-2-ylidene]pyridine-4-carbohydrazide. The resulting product was determined to be polymorphic, exhibiting two crystal forms: form I and form II. From the crystal structure determination using SC-XRD it was determined that form I crystalizes in the C2/c space group while form II crystalizes in the P21/c space group. The hydrogen bonding patterns of both forms are distinctively different from each other: form I forms a chain hydrogen bond motif by forming a hydrogen bond between the hydroxyl group and the oxygen of the amide group while form II forms dimers with a ring hydrogen bond motif forming between the hydroxyl group and the pyridine group. From DSC analysis form I and form II are enantiotropically related, with form I converting to form II at 132.3 °C before melting at 142.3 °C. Based on both experimental and computational evidence, we conclude that form I is a metastable form, with form II being the most stable form. This is another case of a "disappearing polymorph."
Collapse
Affiliation(s)
- Matthew C Scheepers
- Molecular Sciences Institute, School of Chemistry, University of the Witwatersrand Private Bag 3 2050 Johannesburg South Africa +27-11-717-6749 +27-11-717-6711
| | - Manuel A Fernandes
- Molecular Sciences Institute, School of Chemistry, University of the Witwatersrand Private Bag 3 2050 Johannesburg South Africa +27-11-717-6749 +27-11-717-6711
| | - Andreas Lemmerer
- Molecular Sciences Institute, School of Chemistry, University of the Witwatersrand Private Bag 3 2050 Johannesburg South Africa +27-11-717-6749 +27-11-717-6711
| |
Collapse
|
15
|
Loureiro JB, Ribeiro R, Nazareth N, Ferreira T, Lopes EA, Gama A, Machuqueiro M, Alves MG, Marabini L, Oliveira PA, Santos MMM, Saraiva L. Mutant p53 reactivator SLMP53-2 hinders ultraviolet B radiation-induced skin carcinogenesis. Pharmacol Res 2022; 175:106026. [PMID: 34890775 DOI: 10.1016/j.phrs.2021.106026] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/30/2021] [Revised: 12/01/2021] [Accepted: 12/05/2021] [Indexed: 02/07/2023]
Abstract
The growing incidence of skin cancer (SC) has prompted the search for additional preventive strategies to counteract this global health concern. Mutant p53 (mutp53), particularly with ultraviolet radiation (UVR) signature, has emerged as a promising target for SC prevention based on its key role in skin carcinogenesis. Herein, the preventive activity of our previously disclosed mutp53 reactivator SLMP53-2 against UVR-induced SC was investigated. The pre-treatment of keratinocyte HaCaT cells with SLMP53-2, before UVB exposure, depleted mutp53 protein levels with restoration of wild-type-like p53 DNA-binding ability and subsequent transcriptional activity. SLMP53-2 increased cell survival by promoting G1-phase cell cycle arrest, while reducing UVB-induced apoptosis through inhibition of c-Jun N-terminal kinase (JNK) activity. SLMP53-2 also protected cells from reactive oxygen species and oxidative damage induced by UVB. Moreover, it enhanced DNA repair through upregulation of nucleotide excision repair pathway and depletion of UVB-induced DNA damage, as evidenced by a reduction of DNA in comet tails, γH2AX staining and cyclobutane pyrimidine dimers (CPD) levels. SLMP53-2 further suppressed UVB-induced inflammation by inhibiting the nuclear translocation and DNA-binding ability of NF-κB, and promoted the expression of key players involved in keratinocytes differentiation. Consistently, the topical application of SLMP53-2 in mice skin, prior to UVB irradiation, reduced cell death and DNA damage. It also decreased the expression of inflammatory-related proteins and promoted cell differentiation, in UVB-exposed mice skin. Notably, SLMP53-2 did not show signs of skin toxicity for cumulative topical use. Overall, these results support a promising protective activity of SLMP53-2 against UVB-induced SC.
Collapse
Affiliation(s)
- Joana B Loureiro
- LAQV/REQUIMTE, Laboratόrio de Microbiologia, Departamento de Ciências Biolόgicas, Faculdade de Farmácia, Universidade do Porto, 4050-31b Porto, Portugal
| | - Rita Ribeiro
- LAQV/REQUIMTE, Laboratόrio de Microbiologia, Departamento de Ciências Biolόgicas, Faculdade de Farmácia, Universidade do Porto, 4050-31b Porto, Portugal
| | - Nair Nazareth
- LAQV/REQUIMTE, Laboratόrio de Microbiologia, Departamento de Ciências Biolόgicas, Faculdade de Farmácia, Universidade do Porto, 4050-31b Porto, Portugal
| | - Tiago Ferreira
- Centre for Research and Technology of Agro-Environmental and Biological Sciences (CITAB), Inov4Agro, University of Trás-os-Montes and Alto Douro (UTAD), Quinta de Prados, 5000-801 Vila Real, Portugal
| | - Elizabeth A Lopes
- Research Institute for Medicines (iMed.ULisboa), Faculty of Pharmacy, Universidade de Lisboa, 1649-003 Lisboa, Portugal
| | - Adelina Gama
- Animal and Veterinary Research Centre (CECAV), Department of Veterinary Sciences, School of Agrarian and Veterinary Sciences (ECAV), University of Trás-os-Montes and Alto Douro (UTAD), Quinta de Prados, 5000-801 Vila Real, Portugal
| | - Miguel Machuqueiro
- BioISI - Biosystems & Integrative Sciences Institute, Faculty of Sciences, University of Lisboa, Campo Grande, C8 bdg, 1749-016 Lisboa, Portugal
| | - Marco G Alves
- Department of Anatomy and Unit for Multidisciplinary Research in Biomedicine (UMIB), Institute of Biomedical Sciences Abel Salazar (ICBAS), University of Porto, Portugal
| | - Laura Marabini
- Department of Pharmacological and Biomolecular Sciences, Università degli Studi di Milano, Italy
| | - Paula A Oliveira
- Centre for Research and Technology of Agro-Environmental and Biological Sciences (CITAB), Inov4Agro, University of Trás-os-Montes and Alto Douro (UTAD), Quinta de Prados, 5000-801 Vila Real, Portugal
| | - Maria M M Santos
- Research Institute for Medicines (iMed.ULisboa), Faculty of Pharmacy, Universidade de Lisboa, 1649-003 Lisboa, Portugal
| | - Lucília Saraiva
- LAQV/REQUIMTE, Laboratόrio de Microbiologia, Departamento de Ciências Biolόgicas, Faculdade de Farmácia, Universidade do Porto, 4050-31b Porto, Portugal.
| |
Collapse
|