Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 29
Filter
Add more filters










Publication year range
1.
Front Pharmacol ; 15: 1397332, 2024.
Article in English | MEDLINE | ID: mdl-38799161

ABSTRACT

In present times, vanillin stands out as a promising therapeutic molecule that can be implicated in the treatment of neurodegenerative disorders (NDs), notably Alzheimer's disease (AD). This can be attributed to the highly potent scavenging activity of vanillin against reactive oxygen species (ROS). Oxidative stress leads to generation of ROS that serves a critical role in AD's pathological progression. It is apparent from various studies that diets rich in polyphenols prevent oxidative stress associated with AD development, implying the crucial role of vanillin in AD therapeutics. It is crucial to maintain iron balance to manage AD associated oxidative stress, unveiling the significance of human transferrin (hTf) that maintains iron homeostasis. Here, we have performed an integrated study of spectroscopic and computational approaches to get insight into the binding mechanism of vanillin with hTf. In the preliminary study, molecular docking deciphered that vanillin primarily occupies the hTf binding pocket, forming multiple interactions with its key residues. Moreover, the binding mechanism was evaluated at an atomistic level employing comprehensive molecular dynamic (MD) simulation. MD analysis demonstrated that binding of vanillin to hTf stabilizes its structure, without inducing any significant alterations in its native conformation. The docked complex was maintained throughout the simulations without changing its original conformation. Essential dynamics analysis further confirms that hTf achieved a stable conformation with vanillin. The outcomes were further supplemented by fluorescence spectroscopy which confirms the formation of stable hTf-vanillin complex. Taken together, the current study unveils the interaction mechanism of vanillin with hTf and providing a platform to use vanillin in AD therapeutics in the context of iron homeostasis.

2.
J Alzheimers Dis ; 99(1): 333-343, 2024.
Article in English | MEDLINE | ID: mdl-38701154

ABSTRACT

Background: Neurodegeneration is a term describing an irreversible process of neuronal damage. In recent decades, research efforts have been directed towards deepening our knowledge of numerous neurodegenerative disorders, with a particular focus on conditions such as Alzheimer's disease (AD). Human transferrin (htf) is a key player in maintaining iron homeostasis within brain cells. Any disturbance in this equilibrium gives rise to the emergence of neurodegenerative diseases and associated pathologies, particularly AD. Limonene, a natural compound found in citrus fruits and various plants, has shown potential neuroprotective properties. Objective: In this study, our goal was to unravel the binding of limonene with htf, with the intention of comprehending the interaction mechanism of limonene with htf. Methods: Binding was scrutinized using fluorescence quenching and UV-Vis spectroscopic analyses. The binding mechanism of limonene was further investigated at the atomic level through molecular docking and extensive 200 ns molecular dynamic simulation (MD) studies. Results: Molecular docking uncovered that limonene interacted extensively with the deep cavity located within the htf binding pocket. MD results indicated that binding of limonene to htf did not induce substantial structural alterations, ultimately forming stable complex. The findings from fluorescence binding indicated a pronounced interaction between limonene and htf, limonene binds to htf with a binding constant (K) of 0.1×105 M-1. UV spectroscopy also advocated stable htf-limonene complex formation. Conclusions: The study deciphered the binding mechanism of limonene with htf, providing a platform to use limonene in AD therapeutics in context of iron homeostasis.


Subject(s)
Alzheimer Disease , Limonene , Molecular Docking Simulation , Transferrin , Limonene/pharmacology , Limonene/metabolism , Limonene/chemistry , Humans , Alzheimer Disease/metabolism , Alzheimer Disease/drug therapy , Transferrin/metabolism , Molecular Dynamics Simulation , Terpenes/pharmacology , Terpenes/chemistry , Terpenes/metabolism , Protein Binding
3.
Int J Biol Macromol ; 267(Pt 1): 131573, 2024 May.
Article in English | MEDLINE | ID: mdl-38614188

ABSTRACT

DNA, vital for biological processes, encodes hereditary data for protein synthesis, shaping cell structure and function. Since revealing its structure, DNA has become a target for various therapeutically vital molecules, spanning antidiabetic to anticancer drugs. These agents engage with DNA-associated proteins, DNA-RNA hybrids, or bind directly to the DNA helix, triggering diverse downstream effects. These interactions disrupt vital enzymes and proteins essential for maintaining cell structure and function. Analysing drug-DNA interactions has significantly advanced our understanding of drug mechanisms. Glipizide, an antidiabetic drug, is known to cause DNA damage in adipocytes. However, its extract mechanism of DNA interaction is unknown. This study delves into the interaction between glipizide and DNA utilizing various biophysical tools and computational technique to gain insights into the interaction mechanism. Analysis of UV-visible and fluorescence data reveals the formation of complex between DNA and glipizide. The binding affinity of glipizide to DNA was of moderate strength. Examination of thermodynamic parameters at different temperatures suggests that the binding was entropically spontaneous and energetically favourable. Various experiments such as thermal melting assays, viscosity measurement, and dye displacement assays confirmed the minor grove nature of binding of glipizide with DNA. Molecular dynamics studies confirmed the glipizide forms stable complex with DNA when simulated by mimicking the physiological conditions. The binding was mainly favoured by hydrogen bonds and glipizide slightly reduced nucleotide fluctuations of DNA. The study deciphers the mechanism of interaction of glipizide with DNA at molecular levels.


Subject(s)
DNA , Glipizide , Molecular Dynamics Simulation , Thermodynamics , Glipizide/chemistry , Glipizide/pharmacology , DNA/chemistry , DNA/metabolism , Computational Biology/methods , Molecular Docking Simulation , Nucleic Acid Conformation , Hypoglycemic Agents/chemistry , Hypoglycemic Agents/pharmacology
4.
ACS Omega ; 9(14): 16089-16096, 2024 Apr 09.
Article in English | MEDLINE | ID: mdl-38617645

ABSTRACT

Human transferrin (Htf) is vital in maintaining iron within the brain cells; any disruption results in the development of neurodegenerative diseases (NDs) and other related pathologies, especially Alzheimer's disease (AD). Ellagic acid (EA), a naturally occurring phenolic antioxidant, possesses neuroprotective potential and is present in a broad variety of fruits and vegetables. The current work explores the binding mechanism of dietary polyphenol, EA, with Htf by a combination of experimental and computational approaches. Molecular docking studies unveiled the binding of EA to Htf with good affinity. Molecular dynamic (MD) simulation further provided atomistic details of the binding process, demonstrating a stable Htf-EA complex formation without causing substantial alterations to the protein's conformation. Furthermore, fluorescence binding measurements indicated that EA forms a high-affinity interaction with Htf. Isothermal titration calorimetric measurements advocated the spontaneous nature of binding and also revealed the binding process to be exothermic. In conclusion, the study deciphered the binding mechanism of EA with Htf. The results demonstrated that EA binds with Htf with an excellent affinity spontaneously, thereby laying the groundwork for potential applications of EA in the realm of therapeutics for NDs in the context of iron homeostasis.

5.
Int J Biol Macromol ; 259(Pt 2): 129167, 2024 Feb.
Article in English | MEDLINE | ID: mdl-38176507

ABSTRACT

Apolipoprotein E (ApoE), a pivotal contributor to lipid metabolism and neurodegenerative disorders, emerges as an attractive target for therapeutic intervention. Within this study, we deployed an integrated in-silico strategy, harnessing structure-based virtual screening, to identify potential compounds from DrugBank database. Employing molecular docking, we unveil initial hits by evaluating their binding efficiency with ApoE. This first tier of screening narrows our focus to compounds that exhibit a strong propensity to bind with ApoE. Further, a detailed interaction analysis was carried out to explore the binding patterns of the selected hits towards the ApoE binding site. The selected compounds were then evaluated for the biological properties in PASS analysis, which showed anti-neurodegenerative properties. Building upon this foundation, we delve deeper, employing all-atom molecular dynamics (MD) simulations extending over an extensive 500 ns. In particular, Ergotamine and Dihydroergocristine emerge as noteworthy candidates, binding to ApoE in a competitive mode. This intriguing binding behavior positions these compounds as potential candidates warranting further analysis in the pursuit of novel therapeutics targeting complex diseases associated with lipid metabolism and neurodegeneration. This approach holds the promise of catalyzing advancements in therapeutic intervention for complex disorders, thereby reporting a meaningful pace towards improved healthcare outcomes.


Subject(s)
Lipid Metabolism , Molecular Dynamics Simulation , Molecular Docking Simulation , Computational Biology , Apolipoproteins E
6.
Int J Biol Macromol ; 254(Pt 3): 127913, 2024 Jan.
Article in English | MEDLINE | ID: mdl-37939772

ABSTRACT

This is the first study that explored the potential use of Zizyphus mauritiana seed extract (ZSE) to synthesize nano-fluorohydroxyapatite/carboxymethyl chitosan nanocomposite scaffolds at different concentrations (CFZ1, CFZ2 and CFZ3) using co-precipitation method. The proposed scaffolds showed presence of intermolecular H bonding interactions between the constituents, according to the FTIR. The mechanical studies revealed shore hardness of 72 ± 4.6 and optimal compressive modulus in case of CFZ3 [1654.48 ± 1.6 MPa], that was comparable with that of human cortical bone. The SEM, TEM and platelet adhesion images corroborated uniformly distributed needle like particles in case of CFZ3 with an average size ranging from 22 to 26 nm, linked rough morphology, and appropriate hemocompatibility. The markedly up regulation in the ALP activity and protein adsorption upon increasing ZSE concentration demonstrated that CFZ nanocomposite scaffolds were compatible with osteoblastic cells relative to CF nanocomposite. The cytotoxicity study indicated that CFZ nanocomposite do not induce toxicity over MG-63 and did not aggravate LDH leakage in contrast to CF. The histopathological investigations on albino rats confirmed significantly improved regeneration of bone in the repair of a critical-size [8 mm] calvarium defect. Therefore, CFZ3 nanocomposite scaffold represents a simple, off-the-shelf solution to the combined challenges associated with bone defects.


Subject(s)
Chitosan , Nanocomposites , Ziziphus , Rats , Animals , Humans , Tissue Scaffolds , Tissue Engineering/methods , Bone Regeneration , Durapatite/pharmacology
7.
Int J Biol Macromol ; 258(Pt 2): 128900, 2024 Feb.
Article in English | MEDLINE | ID: mdl-38128802

ABSTRACT

Neurological disorders (NDs) have become a major cause of both cognitive and physical disabilities worldwide. In NDs, misfolded proteins tend to adopt a ß-sheet-rich fibrillar structure called amyloid. Amyloid beta (Aß) plays a crucial role in the nervous system. The misfolding and aggregation of Aß are primary factors in the progression of Alzheimer's disease (AD). Inhibiting the oligomerization and aggregation of Aß is considered as an effective strategy against NDs. While it is known that berberine analogs exhibit anti-Aß aggregation properties, the precise mechanism of action remains unclear. In this study, we have employed computational approaches to unravel the possible mechanism by which berberine combats Aß aggregation. The introduction of berberine was observed to delay the equilibrium of Aß16-21 oligomerization. Initially, within the first 10 ns of simulation, ß-sheets content was 12.89 % and gradually increased to 22.19 % within the first 20 ns. This upward trend continued, reaching 32.80 %. However, berberine substantially reduced the formation of ß-sheets to 1.36 %. These findings decipher the potency of berberine against Aß16-21 oligomerization, a crucial step for ß-sheet formation. Additionally, a remarkable decrease in total number of hydrogen bonds was found in the presence of berberine. Berberine also led to a slight reduction in the flexibility of Aß16-21, which may be due to the formation of a more stable structures. This study offers valuable insights at the mechanistic level, which could prove beneficial in the development of new drugs to combat NDs.


Subject(s)
Alzheimer Disease , Berberine , Humans , Amyloid beta-Peptides/metabolism , Berberine/pharmacology , Amyloid/chemistry , Computer Simulation , Molecular Dynamics Simulation , Peptide Fragments/chemistry
8.
Amino Acids ; 55(12): 1923-1935, 2023 Dec.
Article in English | MEDLINE | ID: mdl-37926707

ABSTRACT

Disruptions to iron metabolism and iron homeostasis have emerged as significant contributors to the development and progression of Alzheimer's disease (AD). Human transferrin plays a key part in maintaining iron equilibrium throughout the body, highlighting its importance in AD. Many plant-derived compounds and dietary constituents show promise for preventing AD. Polyphenols that are abundant in fruits, vegetables, teas, coffee, and herbs possess neuroprotective attributes. Resveratrol is a natural polyphenol present in various plant sources like grapes, berries, peanuts, and red wine that has garnered research interest due to its wide range of biological activities. Notably, resveratrol exhibits neuroprotective effects that may help prevent or treat AD through multiple mechanisms. In the present study, we employed a combination of molecular docking and all-atom molecular dynamic simulations (MD) along with experimental approaches to unravel the intricate interactions between transferrin and resveratrol deciphering the binding mechanism. Through molecular docking analysis, it was determined that resveratrol occupies the iron binding pocket of transferrin. Furthermore, MD simulations provided a more profound insight into the stability and conformational dynamics of the complex suggesting that the binding of resveratrol introduced localized flexibility, while maintaining overall stability. The spectroscopic observations yielded clear evidence of substantial binding between resveratrol and transferrin, confirming the computational findings. The identified binding mechanism and conformational stability hold potential for advancing the development of innovative therapeutic approaches targeting AD through resveratrol, particularly concerning iron homeostasis. These insights serve as a platform for considering the natural compounds in the realm of AD therapeutics.


Subject(s)
Alzheimer Disease , Humans , Resveratrol/pharmacology , Resveratrol/therapeutic use , Alzheimer Disease/drug therapy , Alzheimer Disease/metabolism , Transferrin , Molecular Docking Simulation , Polyphenols , Iron/metabolism
9.
Int J Biol Macromol ; 253(Pt 2): 126684, 2023 Dec 31.
Article in English | MEDLINE | ID: mdl-37666395

ABSTRACT

Superoxide dismutase 1 (SOD1) is a vital enzyme responsible for controlling cellular oxidative stress. Any dysregulation of SOD1 activity is linked with cancer pathogenesis and neurodegenerative disorders, such as amyotrophic lateral sclerosis (ALS). Among the inhibitors known to be effective against SOD1, LCS-1 stands out; however, its efficacy, specificity, and safety profiles are somewhat restricted. In this study, we used PubChem library to retrieve compounds that exhibited a structural similarity of at least 90 % with LCS-1. These compounds underwent molecular docking analyses to examine their interaction patterns and binding affinities with SOD1. Further, we applied filters based on physicochemical and ADMET properties, refining the selection process. Our analysis revealed that selected compounds interact with crucial residues of SOD1 active site. To gain further insights into conformational stability and dynamics of the SOD1-ligand complexes, we conducted all-atom molecular dynamics (MD) simulations for 100 ns. We identified two compounds, CID:133306073 and CID:133446715, as potential scaffolds with promising inhibitory properties against SOD1. Both compounds hold significant potential for further exploration as therapeutic SOD1 inhibitors. Further studies are warranted to fully harness their therapeutic potential in targeting SOD1 for cancer and ALS treatment, offering new avenues for improved patient outcomes and disease management.


Subject(s)
Amyotrophic Lateral Sclerosis , Neoplasms , Humans , Superoxide Dismutase-1/genetics , Molecular Docking Simulation , Amyotrophic Lateral Sclerosis/metabolism , Oxidation-Reduction , Superoxide Dismutase/metabolism , Mutation
10.
Int J Biol Macromol ; 253(Pt 1): 126643, 2023 Dec 31.
Article in English | MEDLINE | ID: mdl-37657585

ABSTRACT

Neurodegeneration, a process of irreversible neuronal damage, is characterized by a damaged neuronal structure and function. The interplay between various proteins maintains homeostasis of essential metals in the brain, shielding neurons from degeneration; human transferrin (Htf) is essential in maintaining iron homeostasis. Any disruption in iron homeostasis results in the development of neurodegenerative diseases (NDs) and their pathology, mainly Alzheimer's disease (AD). Rutin is a known compound for its neuroprotective effects. In this work, we deciphered the binding of rutin with Htf in a bid to understand the interaction mechanism. The results of fluorescence and UV-vis spectroscopy demonstrated strong interaction between rutin and Htf. The enthalpy change (∆H°) and entropy change (∆S°) analysis demonstrated hydrophobic interactions as the prevalent forces. The binding mechanism of rutin was further assessed atomistically by molecular docking and extensive 200 ns molecular dynamic simulation (MD) studies; molecular docking showed binding of rutin within Htf's binding pocket. MD results suggested that binding of rutin to Htf does not cause significant structural switching or disruption of the protein's native packing. Overall, the study deciphers the binding of rutin with hTf, delineating the binding mechanism and providing a platform to use rutin in NDs therapeutics.


Subject(s)
Neurodegenerative Diseases , Transferrin , Humans , Transferrin/chemistry , Molecular Docking Simulation , Protein Binding , Neurodegenerative Diseases/drug therapy , Rutin/pharmacology , Iron/chemistry
12.
Int J Biol Macromol ; 245: 125466, 2023 Aug 01.
Article in English | MEDLINE | ID: mdl-37348582

ABSTRACT

Sphingosine kinase 1 (SphK1) has been widely recognized as a significant contributor to various types of cancer, including breast, lung, prostate, and hematological cancers. This research aimed to find a potential SphK1 inhibitor through a step-by-step virtual screening of PF543 (a known SphK1 inhibitor)-like compounds obtained from the PubChem library with the Tanimoto threshold of 80 %. The virtual screening process included several steps, namely physicochemical and ADMET evaluation, PAINS filtering, and molecular docking, followed by molecular dynamics (MD) simulation and principal component analysis (PCA). The results showed that compound CID:58293960 ((3R)-1,1-dioxo-2-[[3-[(4-phenylphenoxy)methyl]phenyl]methyl]-1,2-thiazolidine-3-carboxylic acid) demonstrated high potential as SphK1 inhibitor. All-atom MD simulations were performed for 100 ns to evaluate the stability and structural changes of the docked complexes in an aqueous environment. The analysis of the time evolution data of structural deviations, compactness, PCA, and free energy landscape (FEL) indicated that the binding of CID:58293960 with SphK1 is relatively stable throughout the simulation. The results of this study provide a platform for the discovery and development of new anticancer therapeutics targeting SphK1.


Subject(s)
Molecular Dynamics Simulation , Phosphotransferases (Alcohol Group Acceptor) , Male , Humans , Molecular Docking Simulation , Phosphotransferases (Alcohol Group Acceptor)/chemistry
13.
Int J Biol Macromol ; 237: 124219, 2023 May 15.
Article in English | MEDLINE | ID: mdl-36990415

ABSTRACT

Protein misfolding and related formation of amyloid fibrils are associated with several conformational diseases, such as Alzheimer's disease (AD), Parkinson's disease (PD), Huntington's disease (HD), prion diseases, and Diabetes mellitus, Type 2 (DM-II). Several molecules including antibiotics, polyphenols, flavonoids, anthraquinones, and other small molecules are implicated to modulate amyloid assembly. The stabilization of the native forms of the polypeptides and prevention of their misfolding and aggregation are of clinical and biotechnological importance. Among the natural flavonoids, luteolin is of great importance because of its therapeutic role against neuroinflammation. Herein, we have explored the inhibitory effect of luteolin (LUT) on aggregation of a model protein, human insulin (HI). To understand the molecular mechanism of the inhibition of aggregation of HI by LUT, we employed molecular simulation, UV-Vis, fluorescence, and circular dichroism (CD) spectroscopies along with the dynamic light scattering (DLS). The analysis of the tuning of the HI aggregation process by luteolin revealed that interaction of HI with LUT resulted in the decrease in binding of the various fluorescent dyes, such as thioflavin T (ThT) and 8-anilinonaphthalene-1-sulfonic acid (ANS) to this protein. Retention of the native-like CD spectra and resistance to the aggregation in the presence of LUT has confirmed the aggregation inhibitory potential of LUT. The maximum inhibitory effect was found at the protein-to-drug ratio of 1:12, and no significant change was observed beyond this concentration.


Subject(s)
Amyloidogenic Proteins , Luteolin , Humans , Amyloid/chemistry , Insulin/chemistry , Peptides
14.
Int J Biol Macromol ; 203: 292-301, 2022 Apr 01.
Article in English | MEDLINE | ID: mdl-35074335

ABSTRACT

The present article reports the biogenic synthesis of silver nanoparticles (AgNPs) and gold nanoparticles (AuNPs) from the extract of Acacia auriculiformis (AA) leaves using biogenic approach. Several spectral and morphological studies namely UV-vis, Fourier transform infrared (FT-IR), tunneling electron microscopy along with selected area electron diffraction (TEM/SAED), scanning electron microscopy along with energy dispersive X-ray (SEM-EDX) and X-ray diffraction (XRD) were carried out which ascertains the successful formation of silver nanoparticles (AgNPs) and gold nanoparticles (AuNPs) starting from Silver nitrate and Chloroauric acid respectively. On the basis of TEM/SAED and SEM-EDX, AgNPs were found to be more regular with smaller particle size and hence they were selected for biological studies. Thermal techniques like thermo gravimetric analysis (TGA) and differential thermal analysis (DTA) were also performed to study the comparative thermal stability of AgNPs and AuNPs where AgNPs were found to be thermally more stable. Several biophysical techniques including Thioflavin T assay, ANS assay, Rayleigh scattering method and turbidity assay were also performed. These assays confirm that AgNPs possess better inhibitory property. Moreover, antioxidant activity of AgNPs was also carried out using 2,2-diphenyl-1-picrylhydrazyl (DPPH) and AgNPs were found to be good antioxidant.


Subject(s)
Acacia , Alzheimer Disease , Metal Nanoparticles , Parkinson Disease , Anti-Bacterial Agents/pharmacology , Gold/chemistry , Metal Nanoparticles/chemistry , Plant Extracts/chemistry , Plant Leaves/chemistry , Silver/analysis , Spectroscopy, Fourier Transform Infrared , X-Ray Diffraction
15.
Int J Biol Macromol ; 190: 44-55, 2021 Nov 01.
Article in English | MEDLINE | ID: mdl-34480905

ABSTRACT

With varying clinical symptoms, most neurodegenerative diseases are associated with abnormal loss of neurons. They share the same common pathogenic mechanisms involving misfolding and aggregation, and these visible aggregates of proteins are deposited in the central nervous system. Amyloid formation is thought to arise from partial unfolding of misfolded proteins leading to the exposure of hydrophobic surfaces, which interact with other similar structures and give rise to form dimers, oligomers, protofibrils, and eventually mature fibril aggregates. Accumulating evidence indicates that amyloid oligomers, not amyloid fibrils, are the most toxic species that causes Alzheimer's disease (AD) and Parkinson's disease (PD). AD has recently been recognized as the 'twenty-first century plague', with an incident rate of 1% at 60 years of age, which then doubles every fifth year. Currently, 5.3 million people in the US are afflicted with this disease, and the number of cases is expected to rise to 13.5 million by 2050. PD, a disorder of the brain, is the second most common form of dementia, characterized by difficulty in walking and movement. Keeping the above views in mind, in this review we have focused on the roles of amyloid in neurodegenerative diseases including AD and PD, the involvement of amyloid in mitochondrial dysfunction leading to neurodegeneration, are also considered in the review.


Subject(s)
Alzheimer Disease/metabolism , Amyloid/metabolism , Parkinson Disease/metabolism , Amyloid/chemistry , Animals , Humans , Mitochondria/pathology , Nerve Degeneration/pathology
16.
Int J Biol Macromol ; 186: 580-590, 2021 Sep 01.
Article in English | MEDLINE | ID: mdl-34271045

ABSTRACT

Protein misfolding and aggregation can be induced by a wide variety of factors, such as dominant disease-associated mutations, changes in the environmental conditions (pH, temperature, ionic strength, protein concentration, exposure to transition metal ions, exposure to toxins, posttranslational modifications including glycation, phosphorylation, and sulfation). Misfolded intermediates interact with similar intermediates and progressively form dimers, oligomers, protofibrils, and fibrils. In amyloidoses, fibrillar aggregates are deposited in the tissues either as intracellular inclusion or extracellular plaques (amyloid). When such proteinaceous deposit occurs in the neuronal cells, it initiates degeneration of neurons and consequently resulting in the manifestation of various neurodegenerative diseases. Several different types of molecules have been designed and tested both in vitro and in vivo to evaluate their anti-amyloidogenic efficacies. For instance, the native structure of a protein associated with amyloidosis could be stabilized by ligands, antibodies could be used to remove plaques, oligomer-specific antibody A11 could be used to remove oligomers, or prefibrillar aggregates could be removed by affibodies. Keeping the above views in mind, in this review we have discussed protein misfolding and aggregation, mechanisms of protein aggregation, factors responsible for aggregations, and strategies for aggregation inhibition.


Subject(s)
Alzheimer Disease/drug therapy , Amyloidogenic Proteins/metabolism , Amyloidosis/drug therapy , Nanoparticles , Protein Aggregates , Protein Aggregation, Pathological , Single-Domain Antibodies/pharmacology , Alzheimer Disease/metabolism , Alzheimer Disease/pathology , Amyloidogenic Proteins/antagonists & inhibitors , Amyloidosis/metabolism , Amyloidosis/pathology , Animals , Humans , Protein Conformation , Protein Folding
17.
Curr Protein Pept Sci ; 21(6): 573-583, 2020.
Article in English | MEDLINE | ID: mdl-32013844

ABSTRACT

Protein folding is a natural phenomenon through which a linear polypeptide possessing necessary information attains three-dimension functionally active conformation. This is a complex and multistep process and therefore, the presence of several intermediary structures could be speculated as a result of protein folding. In in vivo, this folding process is governed by the assistance of other proteins called molecular chaperones and heat shock proteins. Due to the mechanism of protein folding, these intermediary structures remain major challenge for modern biology. Mutation in gene encoding amino acid can cause adverse environmental conditions which may result in misfolding of the linear polypeptide followed by the formation of aggregates and amyloidosis. Aggregation contributes to the pathophysiology of several maladies including diabetes mellitus, Huntington's and Alzheimer's disease. The propensity of native structure to form aggregated and fibrillar assemblies is a hallmark of amyloidosis. During aggregation of a protein, transition from α helix to ß sheet is observed, and mainly ß sheeted structure is visualised in a mature fibril. Heme proteins are very crucial for major life activities like transport of oxygen and carbon dioxide, synthesis of ATP, role in electron transport chain, and detoxification of free radicals formed during biochemical reactions. Any structural variation in the heme proteins may lead to a fatal response. Hence characterization of the folding intermediates becomes crucial. The characterization has been deciphered with the help of strong denaturants like acetonitrile and TFE. Moreover, possible role of elimination of these aggregates and prevention of protein denaturation is also discussed. Current review deals with the basic process and mechanism of the protein folding in general and the ultimate outcomes of the protein misfolding. Since Native conformation of heme proteins is essential for some vital activities as listed above, we have discussed possible prevention of denaturation and aggregation of heme proteins such as Hb, cyt c, catalase & peroxidase.


Subject(s)
Alzheimer Disease/metabolism , Amyloid/chemistry , Amyloidosis/metabolism , Diabetes Mellitus/metabolism , Hemoglobins/chemistry , Huntington Disease/metabolism , Alzheimer Disease/genetics , Alzheimer Disease/pathology , Amyloid/genetics , Amyloid/metabolism , Amyloidosis/genetics , Amyloidosis/pathology , Catalase/chemistry , Catalase/genetics , Catalase/metabolism , Cytochromes c/chemistry , Cytochromes c/genetics , Cytochromes c/metabolism , Diabetes Mellitus/genetics , Diabetes Mellitus/pathology , Gene Expression , Heat-Shock Proteins/genetics , Heat-Shock Proteins/metabolism , Hemoglobins/genetics , Hemoglobins/metabolism , Humans , Huntington Disease/genetics , Huntington Disease/pathology , Molecular Chaperones/genetics , Molecular Chaperones/metabolism , Peroxidase/chemistry , Peroxidase/genetics , Peroxidase/metabolism , Protein Aggregates , Protein Conformation , Protein Folding
18.
Int J Biol Macromol ; 135: 337-343, 2019 Aug 15.
Article in English | MEDLINE | ID: mdl-31128176

ABSTRACT

Since aggregation of protein result into number of human diseases including diabetes mellitus, Huntington's and Alzheimer's disease, etc. Hence prevention of aggregation of a polypeptide is of great clinical importance. Human serum albumin (HSA) being major transporter serum protein was studied here in order to prevent its aggregation under extreme conditions. Sulfamethoxazole (SMZ) which is an antibiotic, caused significant inhibition of aggregation which was evident by number of biophysical techniques. Molecular docking was performed to elucidate the protein ligand binding site. In the presence of SMZ decrease in ThT, ANS and RLS fluorescence intensity suggested the inhibitory potency of this antibiotic. Further resistance to increment in the absorbance of Congo red and turbidity was observed even at elevated temperature. Circular dichroism also corroborated these results in retaining its secondary structure in the presence of SMZ. Finally the formation of aggregates, visualized under transmission electron microscopy (TEM) validated the inhibitory tendency of SMZ. Also in the parallel sets we have monitored aggregation kinetics using ThT and turbidity assay and it is noteworthy that SMZ caused maximum inhibition at protein SMZ concentration ratio of 1:30 and 1:40. Our findings would set a hallmark for designing new therapeutics for untreatable protein conformational disorders.


Subject(s)
Anti-Bacterial Agents/chemistry , Peptides/chemistry , Serum Albumin, Human/chemistry , Sulfamethoxazole/chemistry , Humans , Kinetics , Molecular Docking Simulation , Molecular Dynamics Simulation , Molecular Structure , Protein Aggregates , Protein Binding , Protein Conformation , Protein Stability , Protein Structure, Secondary , Spectrum Analysis , Temperature
19.
Int J Biol Macromol ; 129: 1015-1023, 2019 May 15.
Article in English | MEDLINE | ID: mdl-30794897

ABSTRACT

Protein misfolding diseases are associated with human pathologies. These neurodegenerative diseases remain challenging task for researchers because of their adverse effect on vital organs system. Lysozyme amyloidosis is also associated with multi-organ dysfunction. Hence elucidation of its folding pathway is of great importance, for which hen egg white lysozyme (HEWL) being homological to its human counterpart was taken into consideration. Here in this study we have investigated the effect of diosmin (DSN), a flavonoid over thermally aggregated HEWL. Decrease in ANS, ThT and Rayleigh scattering fluorescence intensity suggests the transition between ß to α conformations. Further decrease in absorbance at 360 nm and of congo red with slight blue shift also indicated the disappearance of ß sheeted structure under the under the influence of increasing concentration of DSN. These results were also supported by circular dichroism in which gradual appearance α helical structure was observed. Finally visualization under transmission electron microscopy (TEM) authenticated the maximum structural alteration in the previously formed aggregates of HEWL at 250 µM DSN. Molecular docking followed by 100 ns MD simulations help to understand the interaction mechanism of HEWL with DSN. Results suggest DSN could be a useful in the treatment of amyloid related disorders.


Subject(s)
Amyloid/chemistry , Diosmin/pharmacology , Muramidase/chemistry , Protein Aggregates/drug effects , Protein Unfolding/drug effects , Temperature , Diosmin/metabolism , Molecular Docking Simulation , Molecular Dynamics Simulation , Muramidase/metabolism , Protein Conformation, beta-Strand/drug effects
20.
J Biomol Struct Dyn ; 36(3): 551-560, 2018 02.
Article in English | MEDLINE | ID: mdl-28150569

ABSTRACT

Catalase, a ubiquitous enzyme of the free radical scavenging machinery unfolds and aggregates in the presence of 2,2,2-triflouroethanol (TFE). Catalase molecule aggregates at 50% TFE as evident by high thioflavin T fluorescence, shifted congo red absorbance, change in circular dichroism and soret spectra. TEM images confirmed the nature of catalase aggregates to be oligomers. Organic solvent-induced aggregation of catalase is prevented by the presence of peroxidase (another enzyme of the free radical scavenging machinery). To alter the progress of aggregation in presence of increasing concentration of TFE, we determined the effect of peroxidase on catalase oligomerization by several different techniques, including turbidity measurement, activity assay, thioflavin T fluorescence, circular dichroism, shift in congo red absorbance, transmission electron microscopy (TEM), Rayleigh scattering, soret absorption spectra, and ANS fluorescence. The presence of peroxidase in the vicinity of folded catalase helps it to remain functionally active and inhibited aggregation in the presence of TFE, suggesting that proteins are stable in crowded environments. Moreover, this catalase-peroxidase interaction is biologically significant as it provides insights into how the aggregation process may be altered.


Subject(s)
Catalase/chemistry , Peroxidase/chemistry , Protein Aggregates/drug effects , Protein Multimerization/drug effects , Circular Dichroism , Congo Red , Ethylene Chlorohydrin/analogs & derivatives , Ethylene Chlorohydrin/chemistry , Free Radical Scavengers/chemistry , Protein Binding , Protein Conformation/drug effects , Protein Denaturation , Protein Folding , Spectrometry, Fluorescence
SELECTION OF CITATIONS
SEARCH DETAIL
...