1
|
Rosiles-Alanis W, Zamilpa A, García-Macedo R, Zavala-Sánchez MA, Hidalgo-Figueroa S, Mora-Ramiro B, Román-Ramos R, Estrada-Soto SE, Almanza-Perez JC. 4-Hydroxybenzoic Acid and β-Sitosterol from Cucurbita ficifolia Act as Insulin Secretagogues, Peroxisome Proliferator-Activated Receptor-Gamma Agonists, and Liver Glycogen Storage Promoters: In Vivo, In Vitro, and In Silico Studies. J Med Food 2022; 25:588-596. [PMID: 35708636 DOI: 10.1089/jmf.2021.0071] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022] Open
Abstract
Insulin secretion and GLUT4 expression are two critical events in glucose regulation. The receptors G-protein-coupled receptor 40 (GPR40) and peroxisome proliferator-activated receptor-gamma (PPARγ) modulate these processes, and they represent potential therapeutic targets for new antidiabetic agent's design. Cucurbita ficifolia fruit is used in traditional medicine for diabetes control. Previous studies demonstrated several effects: a hypoglycemic effect mediated by an insulin secretagogue action, antihyperglycemic effect, and promoting liver glycogen storage. Anti-inflammatory and antioxidant effects were also reported. Moreover, some of its phytochemicals have been described, including d-chiro-inositol. However, to understand these effects integrally, other active principles should be investigated. The aim was to perform a chemical fractionation guided by bioassay to isolate and identify other compounds from C. ficifolia fruit that explain its hypoglycemic action as insulin secretagogue, its antihyperglycemic effect by PPARγ activation, and on liver glycogen storage. Three different preparations of C. ficifolia were tested in vivo. Ethyl acetate fraction derived from aqueous extract showed antihyperglycemic effect in an oral glucose tolerance test and was further fractioned. The insulin secretagogue action was tested in RINm5F cells. For the PPARγ activation, C2C12 myocytes were treated with the fractions, and GLUT4 mRNA expression was measured. Chemical fractionation resulted in the isolation and identification of β-sitosterol and 4-hydroxybenzoic acid (4-HBA), which increased insulin secretion, GLUT4, PPARγ, and adiponectin mRNA expression, in addition to an increase in glycogen storage. 4-HBA exhibited an antihyperglycemic effect, while β-sitosterol showed hypoglycemic effect, confirming the wide antidiabetic related results we found in our in vitro models. An in silico study revealed that 4-HBA and β-sitosterol have potential as dual agonists on PPARγ and GPR40 receptors. Both compounds should be considered in the development of new antidiabetic drug development.
Collapse
Affiliation(s)
- Wendoline Rosiles-Alanis
- Postgraduate degree programme in Experimental Biology, DCBS, Autonomous Metropolitan University-Iztapalapa, Mexico City, Mexico
| | - Alejandro Zamilpa
- Southern Biomedical Research Center (CIBIS), Mexican Social Security Institute, Xochitepec, Mexico
| | - Rebeca García-Macedo
- Medical Investigation Unit in Biochemistry, Specialty Hospital, XXI Century National Medical Center, Mexican Social Security Institute (IMSS), Mexico City, Mexico
| | - Miguel A Zavala-Sánchez
- Biological Systems Dept., DCBS, Autonomous Metropolitan University-Xochimilco, Mexico City, Mexico
| | - Sergio Hidalgo-Figueroa
- CONACyT, IPICYT/Consortium for Research, Innovation and Development for Arid Zones, San Luis Potosí, Mexico
| | - Beatriz Mora-Ramiro
- Health Science Dept., DCBS, Autonomous Metropolitan University-Iztapalapa, Mexico City, Mexico
| | - Rubén Román-Ramos
- Health Science Dept., DCBS, Autonomous Metropolitan University-Iztapalapa, Mexico City, Mexico
| | | | - Julio C Almanza-Perez
- Health Science Dept., DCBS, Autonomous Metropolitan University-Iztapalapa, Mexico City, Mexico
| |
Collapse
|
2
|
Weng L, Chen TH, Zheng Q, Weng WH, Huang L, Lai D, Fu YS, Weng CF. Syringaldehyde promoting intestinal motility with suppressing α-amylase hinders starch digestion in diabetic mice. Biomed Pharmacother 2021; 141:111865. [PMID: 34246193 DOI: 10.1016/j.biopha.2021.111865] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2021] [Revised: 06/24/2021] [Accepted: 06/28/2021] [Indexed: 12/14/2022] Open
Abstract
The antihyperglycemic potential of syringaldehyde has been previously investigated; however, the underlying mechanism remains unclear. In this study, we performed a postprandial glucose test (in vivo) including oral glucose tolerance test (OGTT) and oral starch tolerance test (OSTT) in fructose-induced diabetic mice on a high-fat diet for mimicking type 2 diabetes to explore the hypoglycemic efficacy of syringaldehyde and the underlined molecular involvement of syringaldehyde in a glucose-lowering effect. The results revealed that syringaldehyde dose-dependently suppressed blood glucose in both the OSTT and OGTT when referenced to acarbose and metformin, respectively. Surprisingly, syringaldehyde triggered jejunum motility (ex vivo) via activation of the muscarinic-type acetylcholine receptor. By performing virtual screening with molecular docking, the data showed that syringaldehyde nicely interacted with glucagon-like peptide 1 receptor (GLP-1R), peroxisome proliferator-activated receptor (PPAR), dipeptidyl peptidase-IV (DPP-4), acetylcholine M2 receptor, and acetylcholinesterase. These results showed that syringaldehyde can potentiate intestinal contractility to abolish the α-amylase reaction when concurrently reducing retention time and glucose absorption to achieve a glucose-lowering effect in diabetic mice, suggesting its potential therapeutic benefits with improvement for use as a prophylactic and treatment.
Collapse
Affiliation(s)
- Lebin Weng
- Department of Physiology, School of Basic Medical Science, Xiamen Medical College, Xiamen 361023, Fujian, China.
| | - Ting-Hsu Chen
- Department of Life Science and Institute of Biotechnology, National Dong Hwa University, Hualien 97401, Taiwan.
| | - Qingyan Zheng
- Department of Physiology, School of Basic Medical Science, Xiamen Medical College, Xiamen 361023, Fujian, China.
| | - Wei-Hao Weng
- Department of Pharmacy, China Medical University, Taichung 40402, Taiwan.
| | - Liyue Huang
- Department of Physiology, School of Basic Medical Science, Xiamen Medical College, Xiamen 361023, Fujian, China.
| | - Dong Lai
- Medical Research Center, the Second Affiliated Hospital of Xiamen Medical College, Xiamen 361021, Fujian, China.
| | - Yaw-Syan Fu
- Medical Research Center, the Second Affiliated Hospital of Xiamen Medical College, Xiamen 361021, Fujian, China; Department of Anatomy, School of Basic Medical Science, Xiamen Medical College, Xiamen 361023, Fujian, China.
| | - Ching-Feng Weng
- Department of Physiology, School of Basic Medical Science, Xiamen Medical College, Xiamen 361023, Fujian, China; Medical Research Center, the Second Affiliated Hospital of Xiamen Medical College, Xiamen 361021, Fujian, China.
| |
Collapse
|
3
|
Giacoman-Martínez A, Alarcón-Aguilar FJ, Zamilpa A, Huang F, Romero-Nava R, Román-Ramos R, Almanza-Pérez JC. α-Amyrin induces GLUT4 translocation mediated by AMPK and PPARδ/γ in C2C12 myoblasts. Can J Physiol Pharmacol 2021; 99:935-942. [PMID: 33596122 DOI: 10.1139/cjpp-2021-0027] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
α-Amyrin, a natural pentacyclic triterpene, has an antihyperglycemic effect in mice and dual PPARδ/γ action in 3T3-L1 adipocytes, and potential in the control of type 2 diabetes (T2D). About 80% of glucose uptake occurs in skeletal muscle cells, playing a significant role in insulin resistance (IR) and T2D. Peroxisome-proliferator activated receptors (PPARs), in particular PPARδ and PPARγ, are involved in the regulation of lipids and carbohydrates and, along with adenosine-monophosphate (AMP) - activated protein kinase (AMPK) and protein kinase B (Akt), are implicated in translocation of glucose transporter 4 (GLUT4); however, it is still unknown whether α-amyrin can affect these pathways in skeletal muscle cells. Our objective was to determine the action of α-amyrin in PPARδ, PPARγ, AMPK, and Akt in C2C12 myoblasts. The expression of PPARδ, PPARγ, fatty acid transporter protein (FATP), and GLUT4 was quantified using reverse transcription quantitative PCR and Western blot. α-Amyrin increased these markers along with phospho-AMPK (p-AMPK) but not p-Akt. Molecular docking showed that α-amyrin acts as an AMPK-allosteric activator, and may be related to GLUT4 translocation, as evidenced by confocal microscopy. These data support that α-amyrin could have an insulin-mimetic action in C2C12 myoblasts and should be considered as a bioactive molecule for new multitarget drugs with utility in T2D and other metabolic diseases.
Collapse
Affiliation(s)
- Abraham Giacoman-Martínez
- Laboratorio de Farmacología, Departamento de Ciencias de la Salud, División de Ciencias Biológicas y de la Salud, Universidad Autónoma Metropolitana-Iztapalapa, Ciudad de México, México.,Departamento de Farmacología y Toxicología, Hospital Infantil de México Federico Gómez, Ciudad de México, México
| | - Francisco Javier Alarcón-Aguilar
- Laboratorio de Farmacología, Departamento de Ciencias de la Salud, División de Ciencias Biológicas y de la Salud, Universidad Autónoma Metropolitana-Iztapalapa, Ciudad de México, México
| | - Alejandro Zamilpa
- Departamento de Fitoquímica Farmacológica, Centro de Investigación Biomédica del Sur, Instituto Mexicano del Seguro Social, Xochitepec, Morelos, México
| | - Fengyang Huang
- Departamento de Farmacología y Toxicología, Hospital Infantil de México Federico Gómez, Ciudad de México, México
| | - Rodrigo Romero-Nava
- Laboratorio de Farmacología, Departamento de Ciencias de la Salud, División de Ciencias Biológicas y de la Salud, Universidad Autónoma Metropolitana-Iztapalapa, Ciudad de México, México.,Escuela Superior de Medicina del Instituto Politécnico Nacional, Laboratorio de Señalización Intracelular, Sección de Posgrado, Ciudad de México, México
| | - Rubén Román-Ramos
- Laboratorio de Farmacología, Departamento de Ciencias de la Salud, División de Ciencias Biológicas y de la Salud, Universidad Autónoma Metropolitana-Iztapalapa, Ciudad de México, México
| | - Julio César Almanza-Pérez
- Laboratorio de Farmacología, Departamento de Ciencias de la Salud, División de Ciencias Biológicas y de la Salud, Universidad Autónoma Metropolitana-Iztapalapa, Ciudad de México, México
| |
Collapse
|
4
|
Domínguez-Mendoza EA, Galván-Ciprés Y, Martínez-Miranda J, Miranda-González C, Colín-Lozano B, Hernández-Núñez E, Hernández-Bolio GI, Palomino-Hernández O, Navarrete-Vazquez G. Design, Synthesis, and In Silico Multitarget Pharmacological Simulations of Acid Bioisosteres with a Validated In Vivo Antihyperglycemic Effect. Molecules 2021; 26:799. [PMID: 33557136 PMCID: PMC7913794 DOI: 10.3390/molecules26040799] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2021] [Revised: 01/31/2021] [Accepted: 02/02/2021] [Indexed: 01/16/2023] Open
Abstract
Substituted phenylacetic (1-3), phenylpropanoic (4-6), and benzylidenethiazolidine-2,4-dione (7-9) derivatives were designed according to a multitarget unified pharmacophore pattern that has shown robust antidiabetic activity. This bioactivity is due to the simultaneous polypharmacological stimulation of receptors PPARα, PPARγ, and GPR40 and the enzyme inhibition of aldose reductase (AR) and protein tyrosine phosphatase 1B (PTP-1B). The nine compounds share the same four pharmacophore elements: an acid moiety, an aromatic ring, a bulky hydrophobic group, and a flexible linker between the latter two elements. Addition and substitution reactions were performed to obtain molecules at moderated yields. In silico pharmacological consensus analysis (PHACA) was conducted to determine their possible modes of action, protein affinities, toxicological activities, and drug-like properties. The results were combined with in vivo assays to evaluate the ability of these compounds to decrease glucose levels in diabetic mice at a 100 mg/kg single dose. Compounds 6 (a phenylpropanoic acid derivative) and 9 (a benzylidenethiazolidine-2,4-dione derivative) ameliorated the hyperglycemic peak in a statically significant manner in a mouse model of type 2 diabetes. Finally, molecular dynamics simulations were executed on the top performing compounds to shed light on their mechanism of action. The simulations showed the flexible nature of the binding pocket of AR, and showed that both compounds remained bound during the simulation time, although not sharing the same binding mode. In conclusion, we designed nine acid bioisosteres with robust in vivo antihyperglycemic activity that were predicted to have favorable pharmacokinetic and toxicological profiles. Together, these findings provide evidence that supports the molecular design we employed, where the unified pharmacophores possess a strong antidiabetic action due to their multitarget activation.
Collapse
Affiliation(s)
- Elix Alberto Domínguez-Mendoza
- Facultad de Farmacia, Universidad Autónoma del Estado de Morelos, Cuernavaca, Morelos 62209, Mexico; (E.A.D.-M.); (Y.G.-C.); (J.M.-M.); (C.M.-G.); (B.C.-L.)
| | - Yelzyn Galván-Ciprés
- Facultad de Farmacia, Universidad Autónoma del Estado de Morelos, Cuernavaca, Morelos 62209, Mexico; (E.A.D.-M.); (Y.G.-C.); (J.M.-M.); (C.M.-G.); (B.C.-L.)
| | - Josué Martínez-Miranda
- Facultad de Farmacia, Universidad Autónoma del Estado de Morelos, Cuernavaca, Morelos 62209, Mexico; (E.A.D.-M.); (Y.G.-C.); (J.M.-M.); (C.M.-G.); (B.C.-L.)
| | - Cristian Miranda-González
- Facultad de Farmacia, Universidad Autónoma del Estado de Morelos, Cuernavaca, Morelos 62209, Mexico; (E.A.D.-M.); (Y.G.-C.); (J.M.-M.); (C.M.-G.); (B.C.-L.)
| | - Blanca Colín-Lozano
- Facultad de Farmacia, Universidad Autónoma del Estado de Morelos, Cuernavaca, Morelos 62209, Mexico; (E.A.D.-M.); (Y.G.-C.); (J.M.-M.); (C.M.-G.); (B.C.-L.)
| | - Emanuel Hernández-Núñez
- Cátedra CONACyT, Departamento de Recursos del Mar, Centro de Investigación y de Estudios Avanzados, IPN, Unidad Mérida, Yucatan 97310, Mexico; (E.H.-N.); (G.I.H.-B.)
| | - Gloria I. Hernández-Bolio
- Cátedra CONACyT, Departamento de Recursos del Mar, Centro de Investigación y de Estudios Avanzados, IPN, Unidad Mérida, Yucatan 97310, Mexico; (E.H.-N.); (G.I.H.-B.)
| | - Oscar Palomino-Hernández
- Computational Biomedicine (IAS-5/INM-9), Forschungszentrum Juelich, 52425 Julich, Germany;
- Department of Chemistry, Rheinisch-Westfälische Technische Hochschule Aachen, 52425 Aachen, Germany
| | - Gabriel Navarrete-Vazquez
- Facultad de Farmacia, Universidad Autónoma del Estado de Morelos, Cuernavaca, Morelos 62209, Mexico; (E.A.D.-M.); (Y.G.-C.); (J.M.-M.); (C.M.-G.); (B.C.-L.)
| |
Collapse
|
5
|
Herrera-Rueda MÁ, Tlahuext H, Paoli P, Giacoman-Martínez A, Almanza-Pérez JC, Pérez-Sánchez H, Gutiérrez-Hernández A, Chávez-Silva F, Dominguez-Mendoza EA, Estrada-Soto S, Navarrete-Vazquez G. Design, synthesis, in vitro, in vivo and in silico pharmacological characterization of antidiabetic N-Boc-l-tyrosine-based compounds. Biomed Pharmacother 2018; 108:670-678. [PMID: 30245467 DOI: 10.1016/j.biopha.2018.09.074] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2018] [Revised: 09/12/2018] [Accepted: 09/12/2018] [Indexed: 11/16/2022] Open
Abstract
In this study, we synthesized five N-Boc-L-tyrosine-based analogues to glitazars. The in vitro effects of compounds 1-5 on protein tyrosine phosphatase 1B (PTP-1B), peroxisome proliferator-activated receptor alpha and gamma (PPARα/γ), glucose transporter type-4 (GLUT-4) and fatty acid transport protein-1 (FATP-1) activation are reported in this paper. Compounds 1 and 3 were the most active in the in vitro PTP-1B inhibition assay, showing IC50s of approximately 44 μM. Treatment of adipocytes with compound 1 increased the mRNA expression of PPARγ and GLUT-4 by 8- and 3-fold, respectively. Moreover, both compounds (1 and 3) also increased the relative mRNA expression of PPARα (by 8-fold) and FATP-1 (by 15-fold). Molecular docking studies were performed in order to elucidate the polypharmacological binding mode of the most active compounds on these targets. Finally, a murine model of hyperglycemia was used to evaluate the in vivo effectiveness of compounds 1 and 3. We found that both compounds are orally active using an exploratory dose of 100 mg/kg, decreasing the blood glucose concentration in an oral glucose tolerance test and a non-insulin-dependent diabetes mellitus murine model. In conclusion, we demonstrated that both molecules showed strong in vitro and in vivo effects and can be considered polypharmacological antidiabetic candidates.
Collapse
Affiliation(s)
| | - Hugo Tlahuext
- Centro de Investigaciones Químicas, Universidad Autónoma del Estado de Morelos, Cuernavaca, Morelos, 62209, Mexico
| | - Paolo Paoli
- Dipartimento di Scienze Biomediche Sperimentali e Cliniche, Sezione di Scienze Biochimiche, Università degli Studi di Firenze, Viale Morgagni 50, 50134, Firenze, Italy
| | - Abraham Giacoman-Martínez
- Laboratorio de Farmacologia, Depto. Ciencias de la Salud, Universidad Autónoma Metropolitana-Iztapalapa, México, D.F. 09340, Mexico
| | - Julio César Almanza-Pérez
- Laboratorio de Farmacologia, Depto. Ciencias de la Salud, Universidad Autónoma Metropolitana-Iztapalapa, México, D.F. 09340, Mexico
| | - Horacio Pérez-Sánchez
- Bioinformatics and High Performance Computing Research Group (BIO-HPC), Computer Engineering Department, Universidad Católica de Murcia (UCAM), E30107, Murcia, Spain
| | | | - Fabiola Chávez-Silva
- Facultad de Farmacia, Universidad Autónoma del Estado de Morelos, Cuernavaca, Morelos 62209, Mexico
| | | | - Samuel Estrada-Soto
- Facultad de Farmacia, Universidad Autónoma del Estado de Morelos, Cuernavaca, Morelos 62209, Mexico
| | - Gabriel Navarrete-Vazquez
- Facultad de Farmacia, Universidad Autónoma del Estado de Morelos, Cuernavaca, Morelos 62209, Mexico.
| |
Collapse
|
6
|
Colín-Lozano B, Estrada-Soto S, Chávez-Silva F, Gutiérrez-Hernández A, Cerón-Romero L, Giacoman-Martínez A, Almanza-Pérez JC, Hernández-Núñez E, Wang Z, Xie X, Cappiello M, Balestri F, Mura U, Navarrete-Vazquez G. Design, Synthesis and in Combo Antidiabetic Bioevaluation of Multitarget Phenylpropanoic Acids. Molecules 2018; 23:molecules23020340. [PMID: 29415496 PMCID: PMC6017591 DOI: 10.3390/molecules23020340] [Citation(s) in RCA: 31] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2018] [Revised: 02/01/2018] [Accepted: 02/03/2018] [Indexed: 12/22/2022] Open
Abstract
We have synthesized a small series of five 3-[4-arylmethoxy)phenyl]propanoic acids employing an easy and short synthetic pathway. The compounds were tested in vitro against a set of four protein targets identified as key elements in diabetes: G protein-coupled receptor 40 (GPR40), aldose reductase (AKR1B1), peroxisome proliferator-activated receptor gama (PPARγ) and solute carrier family 2 (facilitated glucose transporter), member 4 (GLUT-4). Compound 1 displayed an EC50 value of 0.075 μM against GPR40 and was an AKR1B1 inhibitor, showing IC50 = 7.4 μM. Compounds 2 and 3 act as slightly AKR1B1 inhibitors, potent GPR40 agonists and showed an increase of 2 to 4-times in the mRNA expression of PPARγ, as well as the GLUT-4 levels. Docking studies were conducted in order to explain the polypharmacological mode of action and the interaction binding mode of the most active molecules on these targets, showing several coincidences with co-crystal ligands. Compounds 1–3 were tested in vivo at an explorative 100 mg/kg dose, being 2 and 3 orally actives, reducing glucose levels in a non-insulin-dependent diabetes mice model. Compounds 2 and 3 displayed robust in vitro potency and in vivo efficacy, and could be considered as promising multitarget antidiabetic candidates. This is the first report of a single molecule with these four polypharmacological target action.
Collapse
Affiliation(s)
- Blanca Colín-Lozano
- Facultad de Farmacia, Universidad Autónoma del Estado de Morelos, Cuernavaca, Morelos 62209, Mexico.
| | - Samuel Estrada-Soto
- Facultad de Farmacia, Universidad Autónoma del Estado de Morelos, Cuernavaca, Morelos 62209, Mexico.
| | - Fabiola Chávez-Silva
- Facultad de Farmacia, Universidad Autónoma del Estado de Morelos, Cuernavaca, Morelos 62209, Mexico.
| | | | - Litzia Cerón-Romero
- Facultad de Farmacia, Universidad Autónoma del Estado de Morelos, Cuernavaca, Morelos 62209, Mexico.
| | - Abraham Giacoman-Martínez
- Laboratorio de Farmacología, Departamento de Ciencias de la Salud, Universidad Autónoma Metropolitana Iztapalapa, Ciudad de México 09340, Mexico.
| | - Julio Cesar Almanza-Pérez
- Laboratorio de Farmacología, Departamento de Ciencias de la Salud, Universidad Autónoma Metropolitana Iztapalapa, Ciudad de México 09340, Mexico.
| | - Emanuel Hernández-Núñez
- Cátedra CONACyT, Departamento de Recursos del Mar, Centro de Investigación y de Estudios Avanzados del IPN, Unidad Mérida, Yucatán 97310, Mexico.
| | - Zhilong Wang
- CAS Key Laboratory of Receptor Research, the National Center for Drug Screening, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai 201203, China, (Z.W.).
| | - Xin Xie
- CAS Key Laboratory of Receptor Research, the National Center for Drug Screening, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai 201203, China, (Z.W.).
| | - Mario Cappiello
- Dipartimento di Biologia, Unità di Biochimica, University of Pisa, 56126 Pisa, Italy.
| | - Francesco Balestri
- Dipartimento di Biologia, Unità di Biochimica, University of Pisa, 56126 Pisa, Italy.
| | - Umberto Mura
- Dipartimento di Biologia, Unità di Biochimica, University of Pisa, 56126 Pisa, Italy.
| | - Gabriel Navarrete-Vazquez
- Facultad de Farmacia, Universidad Autónoma del Estado de Morelos, Cuernavaca, Morelos 62209, Mexico.
| |
Collapse
|