1
|
Kim TK, Cho Y, Kim J, Lee J, Hong JM, Cho H, Kim JS, Lee Y, Kim KH, Kim IC, Han SJ, Oh H, Jo DG, Yim JH. Synthesis and Evaluation of Chloride-Substituted Ramalin Derivatives for Alzheimer's Disease Treatment. Molecules 2024; 29:3701. [PMID: 39125105 PMCID: PMC11313798 DOI: 10.3390/molecules29153701] [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: 07/09/2024] [Revised: 07/29/2024] [Accepted: 07/29/2024] [Indexed: 08/12/2024] Open
Abstract
Alzheimer's disease (AD) is a progressive neurodegenerative disorder marked by the accumulation of amyloid-beta plaques and hyperphosphorylated tau proteins, leading to cognitive decline and neuronal death. However, despite extensive research, there are still no effective treatments for this condition. In this study, a series of chloride-substituted Ramalin derivatives is synthesized to optimize their antioxidant, anti-inflammatory, and their potential to target key pathological features of Alzheimer's disease. The effect of the chloride position on these properties is investigated, specifically examining the potential of these derivatives to inhibit tau aggregation and beta-site amyloid precursor protein cleaving enzyme 1 (BACE-1) activity. Our findings demonstrate that several derivatives, particularly RA-3Cl, RA-4Cl, RA-26Cl, RA-34Cl, and RA-35Cl, significantly inhibit tau aggregation with inhibition rates of approximately 50%. For BACE-1 inhibition, Ramalin and RA-4Cl also significantly decrease BACE-1 expression in N2a cells by 40% and 38%, respectively, while RA-23Cl and RA-24Cl showed inhibition rates of 30% and 35% in SH-SY5Y cells. These results suggest that chloride-substituted Ramalin derivatives possess promising multifunctional properties for AD treatment, warranting further investigation and optimization for clinical applications.
Collapse
Affiliation(s)
- Tai Kyoung Kim
- Division of Polar Life Sciences, Korea Polar Research Institute, Incheon 21990, Republic of Korea; (T.K.K.); (J.K.); (J.-M.H.); (K.H.K.); (I.-C.K.); (S.J.H.)
| | - Yongeun Cho
- School of Pharmacy, Sungkyunkwan University, Suwon 16419, Republic of Korea; (Y.C.); (J.L.); (H.C.); (J.-S.K.); (Y.L.)
| | - Jaewon Kim
- Division of Polar Life Sciences, Korea Polar Research Institute, Incheon 21990, Republic of Korea; (T.K.K.); (J.K.); (J.-M.H.); (K.H.K.); (I.-C.K.); (S.J.H.)
- Department of Plant Biotechnology, Korea University, Seoul 02841, Republic of Korea
| | - Jeongmi Lee
- School of Pharmacy, Sungkyunkwan University, Suwon 16419, Republic of Korea; (Y.C.); (J.L.); (H.C.); (J.-S.K.); (Y.L.)
| | - Ju-Mi Hong
- Division of Polar Life Sciences, Korea Polar Research Institute, Incheon 21990, Republic of Korea; (T.K.K.); (J.K.); (J.-M.H.); (K.H.K.); (I.-C.K.); (S.J.H.)
| | - Heewon Cho
- School of Pharmacy, Sungkyunkwan University, Suwon 16419, Republic of Korea; (Y.C.); (J.L.); (H.C.); (J.-S.K.); (Y.L.)
| | - Jun-Sik Kim
- School of Pharmacy, Sungkyunkwan University, Suwon 16419, Republic of Korea; (Y.C.); (J.L.); (H.C.); (J.-S.K.); (Y.L.)
| | - Yeongyeong Lee
- School of Pharmacy, Sungkyunkwan University, Suwon 16419, Republic of Korea; (Y.C.); (J.L.); (H.C.); (J.-S.K.); (Y.L.)
| | - Kyung Hee Kim
- Division of Polar Life Sciences, Korea Polar Research Institute, Incheon 21990, Republic of Korea; (T.K.K.); (J.K.); (J.-M.H.); (K.H.K.); (I.-C.K.); (S.J.H.)
- Department of Chemistry, Hanseo University, Seosan 31962, Republic of Korea
| | - Il-Chan Kim
- Division of Polar Life Sciences, Korea Polar Research Institute, Incheon 21990, Republic of Korea; (T.K.K.); (J.K.); (J.-M.H.); (K.H.K.); (I.-C.K.); (S.J.H.)
| | - Se Jong Han
- Division of Polar Life Sciences, Korea Polar Research Institute, Incheon 21990, Republic of Korea; (T.K.K.); (J.K.); (J.-M.H.); (K.H.K.); (I.-C.K.); (S.J.H.)
| | - Hyuncheol Oh
- College of Pharmacy, Wonkwang University, Iksan 54538, Republic of Korea;
| | - Dong-Gyu Jo
- School of Pharmacy, Sungkyunkwan University, Suwon 16419, Republic of Korea; (Y.C.); (J.L.); (H.C.); (J.-S.K.); (Y.L.)
| | - Joung Han Yim
- Division of Polar Life Sciences, Korea Polar Research Institute, Incheon 21990, Republic of Korea; (T.K.K.); (J.K.); (J.-M.H.); (K.H.K.); (I.-C.K.); (S.J.H.)
| |
Collapse
|
2
|
Obaha A, Novinec M. Regulation of Peptidase Activity beyond the Active Site in Human Health and Disease. Int J Mol Sci 2023; 24:17120. [PMID: 38069440 PMCID: PMC10707025 DOI: 10.3390/ijms242317120] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2023] [Revised: 12/01/2023] [Accepted: 12/02/2023] [Indexed: 12/18/2023] Open
Abstract
This comprehensive review addresses the intricate and multifaceted regulation of peptidase activity in human health and disease, providing a comprehensive investigation that extends well beyond the boundaries of the active site. Our review focuses on multiple mechanisms and highlights the important role of exosites, allosteric sites, and processes involved in zymogen activation. These mechanisms play a central role in shaping the complex world of peptidase function and are promising potential targets for the development of innovative drugs and therapeutic interventions. The review also briefly discusses the influence of glycosaminoglycans and non-inhibitory binding proteins on enzyme activities. Understanding their role may be a crucial factor in the development of therapeutic strategies. By elucidating the intricate web of regulatory mechanisms that control peptidase activity, this review deepens our understanding in this field and provides a roadmap for various strategies to influence and modulate peptidase activity.
Collapse
Affiliation(s)
| | - Marko Novinec
- Faculty of Chemistry and Chemical Technology, Department of Chemistry and Biochemistry, University of Ljubljana, Večna pot 113, SI-1000 Ljubljana, Slovenia;
| |
Collapse
|
3
|
Panchal J, Jaiswal S, Jain S, Kumawat J, Sharma A, Jain P, Jain S, Verma K, Dwivedi J, Sharma S. Development of novel bosentan analogues as endothelin receptor antagonists for pulmonary arterial hypertension. Eur J Med Chem 2023; 259:115681. [PMID: 37515921 DOI: 10.1016/j.ejmech.2023.115681] [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: 04/08/2023] [Revised: 07/22/2023] [Accepted: 07/23/2023] [Indexed: 07/31/2023]
Abstract
Since decades, bosentan has been in use for the treatment of pulmonary arterial hypertension (PAH). However, chronic exposure to bosentan leads to the development of resistance, tolerance, and serious adverse effects that have restricted its usage in clinical practices. To surmount these limitations, some new bosentan derivatives have been synthesized and evaluated for their therapeutic efficacy in PAH. Molecular docking analyses of all the synthesized derivatives were carried out using the endothelin (ET) receptor. In addition, the inhibitory ability of synthesized derivatives was determined in in vitro assay employing an ET-1 human ELISA kit. Among the synthesized derivatives, three derivatives namely 17d, 16j, and 16h with higher docking scores and lower IC50 values were selected for determination of the magnitude of the binding force between the derivative and ET receptor using molecular dynamics (MD) simulations study. Further, these derivatives were subjected to in vivo studies using monocrotaline (MCT) induced PAH in rat model. Results of in vivo studies inferred that the derivatives exhibit impressive ability to reduce PAH. Besides, its protective role was also evidenced in hemodynamic and right ventricular hypertrophy analyses, histological analysis, cardiac biomarkers, hypoxia-inducible factor 1 alpha (HIF1α) levels, and biochemical studies. Furthermore, gene quantification by quantitative RT-PCR and Western blot analysis was also performed to examine its effect on the expression of key proteins in PAH. Notably, amongst three, derivative 16h exhibited the most encouraging results in molecular docking analysis, in vitro, in vivo, histopathological, biochemical, protein expression, and MD studies. Besides, derivative 16h also showed impressive pharmacokinetic features in ADMET analysis. In conclusion, derivative 16 h could act as a reliable ET receptor antagonist and requires further exploration to attain its therapeutic utility in PAH management.
Collapse
Affiliation(s)
- Jigar Panchal
- Department of Chemistry, Banasthali Vidyapith, Banasthali, 304022, Rajasthan, India
| | - Shivangi Jaiswal
- Department of Chemistry, Banasthali Vidyapith, Banasthali, 304022, Rajasthan, India
| | - Sonika Jain
- Department of Chemistry, Banasthali Vidyapith, Banasthali, 304022, Rajasthan, India.
| | - Jyoti Kumawat
- Department of Chemistry, Banasthali Vidyapith, Banasthali, 304022, Rajasthan, India
| | - Ashima Sharma
- Department of Pharmacy, Panjab University, Chandigarh, 160014, Punjab, India
| | - Pankaj Jain
- Department of Pharmacy, Banasthali Vidyapith, Banasthali, 304022, Rajasthan, India
| | - Smita Jain
- Department of Pharmacy, Banasthali Vidyapith, Banasthali, 304022, Rajasthan, India
| | - Kanika Verma
- Department of Pharmacy, Banasthali Vidyapith, Banasthali, 304022, Rajasthan, India
| | - Jaya Dwivedi
- Department of Chemistry, Banasthali Vidyapith, Banasthali, 304022, Rajasthan, India.
| | - Swapnil Sharma
- Department of Pharmacy, Banasthali Vidyapith, Banasthali, 304022, Rajasthan, India.
| |
Collapse
|
4
|
Mushebenge AGA, Ugbaja SC, Mbatha NA, B. Khan R, Kumalo HM. Assessing the Potential Contribution of In Silico Studies in Discovering Drug Candidates That Interact with Various SARS-CoV-2 Receptors. Int J Mol Sci 2023; 24:15518. [PMID: 37958503 PMCID: PMC10647470 DOI: 10.3390/ijms242115518] [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: 08/03/2023] [Revised: 10/18/2023] [Accepted: 10/23/2023] [Indexed: 11/15/2023] Open
Abstract
The COVID-19 pandemic has spurred intense research efforts to identify effective treatments for SARS-CoV-2. In silico studies have emerged as a powerful tool in the drug discovery process, particularly in the search for drug candidates that interact with various SARS-CoV-2 receptors. These studies involve the use of computer simulations and computational algorithms to predict the potential interaction of drug candidates with target receptors. The primary receptors targeted by drug candidates include the RNA polymerase, main protease, spike protein, ACE2 receptor, and transmembrane protease serine 2 (TMPRSS2). In silico studies have identified several promising drug candidates, including Remdesivir, Favipiravir, Ribavirin, Ivermectin, Lopinavir/Ritonavir, and Camostat Mesylate, among others. The use of in silico studies offers several advantages, including the ability to screen a large number of drug candidates in a relatively short amount of time, thereby reducing the time and cost involved in traditional drug discovery methods. Additionally, in silico studies allow for the prediction of the binding affinity of the drug candidates to target receptors, providing insight into their potential efficacy. This study is aimed at assessing the useful contributions of the application of computational instruments in the discovery of receptors targeted in SARS-CoV-2. It further highlights some identified advantages and limitations of these studies, thereby revealing some complementary experimental validation to ensure the efficacy and safety of identified drug candidates.
Collapse
Affiliation(s)
- Aganze Gloire-Aimé Mushebenge
- Discipline of Pharmaceutical Sciences, University of KwaZulu-Natal, Westville, Durban 4000, South Africa;
- Drug Research and Innovation Unit, Discipline of Medical Biochemistry, School of Laboratory Medicine and Medical Science, University of KwaZulu-Natal, Durban 4000, South Africa
- Faculty of Pharmaceutical Sciences, University of Lubumbashi, Lubumbashi 1825, Democratic Republic of the Congo
| | - Samuel Chima Ugbaja
- Drug Research and Innovation Unit, Discipline of Medical Biochemistry, School of Laboratory Medicine and Medical Science, University of KwaZulu-Natal, Durban 4000, South Africa
- Africa Health Research Institute, University of KwaZulu-Natal, Durban 4000, South Africa
| | - Nonkululeko Avril Mbatha
- KwaZulu-Natal Research Innovation and Sequencing Platform, School of Laboratory Medicine and Medical Science, University of KwaZulu-Natal, Durban 4000, South Africa
| | - Rene B. Khan
- Drug Research and Innovation Unit, Discipline of Medical Biochemistry, School of Laboratory Medicine and Medical Science, University of KwaZulu-Natal, Durban 4000, South Africa
| | - Hezekiel M. Kumalo
- Drug Research and Innovation Unit, Discipline of Medical Biochemistry, School of Laboratory Medicine and Medical Science, University of KwaZulu-Natal, Durban 4000, South Africa
| |
Collapse
|
5
|
Mushebenge AG, Ugbaja SC, Mtambo SE, Ntombela T, Metu JI, Babayemi O, Chima JI, Appiah-Kubi P, Odugbemi AI, Ntuli ML, Khan R, Kumalo HM. Unveiling the Inhibitory Potentials of Peptidomimetic Azanitriles and Pyridyl Esters towards SARS-CoV-2 Main Protease: A Molecular Modelling Investigation. Molecules 2023; 28:molecules28062641. [PMID: 36985614 PMCID: PMC10051727 DOI: 10.3390/molecules28062641] [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: 04/12/2022] [Revised: 02/14/2023] [Accepted: 03/02/2023] [Indexed: 03/17/2023] Open
Abstract
The severe acute respiratory syndrome coronavirus-2 (SARS-CoV-2) is responsible for COVID-19, which was declared a global pandemic in March 2020 by the World Health Organization (WHO). Since SARS-CoV-2 main protease plays an essential role in the virus’s life cycle, the design of small drug molecules with lower molecular weight has been a promising development targeting its inhibition. Herein, we evaluated the novel peptidomimetic azatripeptide and azatetrapeptide nitriles against SARS-CoV-2 main protease. We employed molecular dynamics (MD) simulations to elucidate the selected compounds’ binding free energy profiles against SARS-CoV-2 and further unveil the residues responsible for the drug-binding properties. Compound 8 exhibited the highest binding free energy of −49.37 ± 0.15 kcal/mol, followed by compound 7 (−39.83 ± 0.19 kcal/mol), while compound 17 showed the lowest binding free energy (−23.54 ± 0.19 kcal/mol). In addition, the absorption, distribution, metabolism, and excretion (ADME) assessment was performed and revealed that only compound 17 met the drug-likeness parameters and exhibited high pharmacokinetics to inhibit CYP1A2, CYP2C19, and CYP2C9 with better absorption potential and blood-brain barrier permeability (BBB) index. The additional intermolecular evaluations suggested compound 8 as a promising drug candidate for inhibiting SARS-CoV-2 Mpro. The substitution of isopropane in compound 7 with an aromatic benzene ring in compound 8 significantly enhanced the drug’s ability to bind better at the active site of the SARS-CoV-2 Mpro.
Collapse
Affiliation(s)
- Aganze G. Mushebenge
- Drug Research and Innovation Unit, Discipline of Medical Biochemistry, School of Laboratory Medicine and Medical Science, University of KwaZulu-Natal, Durban 4000, South Africa; (A.G.M.); (S.E.M.); (J.I.C.); (P.A.-K.); (R.K.)
| | - Samuel C. Ugbaja
- Drug Research and Innovation Unit, Discipline of Medical Biochemistry, School of Laboratory Medicine and Medical Science, University of KwaZulu-Natal, Durban 4000, South Africa; (A.G.M.); (S.E.M.); (J.I.C.); (P.A.-K.); (R.K.)
- Correspondence: (S.C.U.); (H.M.K.)
| | - Sphamandla E. Mtambo
- Drug Research and Innovation Unit, Discipline of Medical Biochemistry, School of Laboratory Medicine and Medical Science, University of KwaZulu-Natal, Durban 4000, South Africa; (A.G.M.); (S.E.M.); (J.I.C.); (P.A.-K.); (R.K.)
| | - Thandokuhle Ntombela
- Catalysis and Peptide Research Unit, School of Pharmaceutical Sciences, University of KwaZulu-Natal, Durban 4000, South Africa;
| | - Joy I. Metu
- National Institute for Nigerian Languages, Aba 453106, Nigeria;
| | - Oludotun Babayemi
- Cloneshouse Nigeria, 6th Floor, Left Wing, NICON Plaza, Plot 242, Muhammadu Buhari Way, Central Business District, Abuja 900103, Nigeria;
| | - Joy I. Chima
- Drug Research and Innovation Unit, Discipline of Medical Biochemistry, School of Laboratory Medicine and Medical Science, University of KwaZulu-Natal, Durban 4000, South Africa; (A.G.M.); (S.E.M.); (J.I.C.); (P.A.-K.); (R.K.)
| | - Patrick Appiah-Kubi
- Drug Research and Innovation Unit, Discipline of Medical Biochemistry, School of Laboratory Medicine and Medical Science, University of KwaZulu-Natal, Durban 4000, South Africa; (A.G.M.); (S.E.M.); (J.I.C.); (P.A.-K.); (R.K.)
| | - Adeshina I. Odugbemi
- South African National Bioinformatics Institute, Faculty of Natural Sciences, University of the Western Cape, Cape Town 7535, South Africa;
| | - Mthobisi L. Ntuli
- Department of Mathematics, Faculty of Applied Science, Durban University of Technology, Durban 4000, South Africa;
| | - Rene Khan
- Drug Research and Innovation Unit, Discipline of Medical Biochemistry, School of Laboratory Medicine and Medical Science, University of KwaZulu-Natal, Durban 4000, South Africa; (A.G.M.); (S.E.M.); (J.I.C.); (P.A.-K.); (R.K.)
| | - Hezekiel M. Kumalo
- Drug Research and Innovation Unit, Discipline of Medical Biochemistry, School of Laboratory Medicine and Medical Science, University of KwaZulu-Natal, Durban 4000, South Africa; (A.G.M.); (S.E.M.); (J.I.C.); (P.A.-K.); (R.K.)
- Correspondence: (S.C.U.); (H.M.K.)
| |
Collapse
|