Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 46
Filtrar
Más filtros

Banco de datos
Tipo del documento
Intervalo de año de publicación
1.
Mol Microbiol ; 120(2): 210-223, 2023 08.
Artículo en Inglés | MEDLINE | ID: mdl-37350285

RESUMEN

Mycobacterium tuberculosis encodes two chaperonin proteins, MtbCpn60.1 and MtbCpn60.2, that share substantial sequence similarity with the Escherichia coli chaperonin, GroEL. However, unlike GroEL, MtbCpn60.1 and MtbCpn60.2 purify as lower-order oligomers. Previous studies have shown that MtbCpn60.2 can functionally replace GroEL in E. coli, while the function of MtbCpn60.1 remained an enigma. Here, we demonstrate the molecular chaperone function of MtbCpn60.1 and MtbCpn60.2, by probing their ability to assist the folding of obligate chaperonin clients, DapA, FtsE and MetK, in an E. coli strain depleted of endogenous GroEL. We show that both MtbCpn60.1 and MtbCpn60.2 support cell survival and cell division by assisting the folding of DapA and FtsE, but only MtbCpn60.2 completely rescues GroEL-depleted E. coli cells. We also show that, unlike MtbCpn60.2, MtbCpn60.1 has limited ability to support cell growth and proliferation and assist the folding of MetK. Our findings suggest that the client pools of GroEL and MtbCpn60.2 overlap substantially, while MtbCpn60.1 folds only a small subset of GroEL clients. We conclude that the differences between MtbCpn60.1 and MtbCpn60.2 may be a consequence of their intrinsic sequence features, which affect their thermostability, efficiency, clientomes and modes of action.


Asunto(s)
Proteínas de Escherichia coli , Mycobacterium tuberculosis , Humanos , Escherichia coli/genética , Escherichia coli/metabolismo , Proteostasis , Chaperoninas/genética , Chaperoninas/metabolismo , Chaperonas Moleculares/metabolismo , Pliegue de Proteína , Mycobacterium tuberculosis/genética , Mycobacterium tuberculosis/metabolismo , Transportadoras de Casetes de Unión a ATP/metabolismo , Regulador de Conductancia de Transmembrana de Fibrosis Quística/metabolismo , Proteínas de Escherichia coli/metabolismo
2.
Biochemistry ; 61(14): 1473-1484, 2022 07 19.
Artículo en Inglés | MEDLINE | ID: mdl-35749234

RESUMEN

Dialysis-related amyloidosis (DRA) is considered an inescapable consequence of renal failure. Upon prolonged hemodialysis, it involves accumulation of toxic ß2-microglobulin (ß2m) amyloids in bones and joints. Current treatment methods are plagued with high cost, low specificity, and low capacity. Through our in vitro and in cellulo studies, we introduce a peptidomimetic-based approach to help develop future therapeutics against DRA. Our study reports the ability of a nontoxic, core-modified, bispidine peptidomimetic analogue "B(LVI)2" to inhibit acid-induced amyloid fibrillation of ß2m (Hß2m). Using thioflavin-T, sodium dodecyl sulfate-polyacrylamide gel electrophoresis, and transmission electron microscopy analysis, we demonstrate that B(LVI)2 delays aggregation lag time of Hß2m amyloid fibrillation and reduces the yield of Hß2m amyloid fibrils in a dose-dependent manner. Our findings suggest a B(LVI)2-orchestrated alteration in the route of Hß2m amyloid fibrillation resulting in the formation of noncytotoxic, morphologically distinct amyloid-like species. Circular dichroism data show gradual sequestration of Hß2m species in a soluble nonamyloidogenic noncytotoxic conformation in the presence of B(LVI)2. Dynamic light scattering measurements indicate incompetence of Hß2m species in the presence of B(LVI)2 to undergo amyloid-competent intermolecular associations. Overall, our study reports the antifibrillation property of a novel peptidomimetic with the potential to bring a paradigm shift in therapeutic approaches against DRA.


Asunto(s)
Amiloidosis , Peptidomiméticos , Amiloide , Proteínas Amiloidogénicas , Amiloidosis/tratamiento farmacológico , Compuestos Bicíclicos Heterocíclicos con Puentes , Humanos , Peptidomiméticos/farmacología , Diálisis Renal , Microglobulina beta-2
3.
Microb Cell Fact ; 21(1): 20, 2022 Feb 05.
Artículo en Inglés | MEDLINE | ID: mdl-35123481

RESUMEN

BACKGROUND: During the recombinant protein expression, most heterologous proteins expressed in E. coli cell factories are generated as insoluble and inactive aggregates, which prohibit E. coli from being employed as an expression host despite its numerous advantages and ease of use. The yeast mitochondrial aconitase protein, which has a tendency to aggregate when expressed in E. coli cells in the absence of heterologous chaperones GroEL/ES was utilised as a model to investigate how the modulation of physiological stimuli in the host cell can increase protein solubility. The presence of folding modulators such as exogenous molecular chaperones or osmolytes, as well as process variables such as incubation temperature, inducer concentrations, growth media are all important for cellular folding and are investigated in this study. This study also investigated how the cell's stress response system activates and protects the proteins from aggregation. RESULTS: The cells exposed to osmolytes plus a pre-induction heat shock showed a substantial increase in recombinant aconitase activity when combined with modulation of process conditions. The concomitant GroEL/ES expression further assists the folding of these soluble aggregates and increases the functional protein molecules in the cytoplasm of the recombinant E. coli cells. CONCLUSIONS: The recombinant E. coli cells enduring physiological stress provide a cytosolic environment for the enhancement in the solubility and activity of the recombinant proteins. GroEL/ES-expressing cells not only aided in the folding of recombinant proteins, but also had an effect on the physiology of the expression host. The improvement in the specific growth rate and aconitase production during chaperone GroEL/ES co-expression is attributed to the reduction in overall cellular stress caused by the expression host's aggregation-prone recombinant protein expression.


Asunto(s)
Aconitato Hidratasa/química , Escherichia coli/metabolismo , Proteínas Reguladoras del Hierro/química , Pliegue de Proteína , Proteínas Recombinantes/química , Aconitato Hidratasa/genética , Aconitato Hidratasa/metabolismo , Escherichia coli/química , Escherichia coli/genética , Escherichia coli/crecimiento & desarrollo , Proteínas Reguladoras del Hierro/genética , Proteínas Reguladoras del Hierro/metabolismo , Chaperonas Moleculares/genética , Chaperonas Moleculares/metabolismo , Proteínas Recombinantes/genética , Proteínas Recombinantes/metabolismo
4.
J Biol Chem ; 294(16): 6450-6467, 2019 04 19.
Artículo en Inglés | MEDLINE | ID: mdl-30792306

RESUMEN

Heat shock protein 90 (Hsp90) is a eukaryotic chaperone responsible for the folding and functional activation of numerous client proteins, many of which are oncoproteins. Thus, Hsp90 inhibition has been intensely pursued, resulting in the development of many potential Hsp90 inhibitors, not all of which are well-characterized. Hsp90 inhibitors not only abrogate its chaperone functions, but also could help us gain insight into the structure-function relationship of this chaperone. Here, using biochemical and cell-based assays along with isothermal titration calorimetry, we investigate KU-32, a derivative of the Hsp90 inhibitor novobiocin (NB), for its ability to modulate Hsp90 chaperone function. Although NB and KU-32 differ only slightly in structure, we found that upon binding, they induce completely opposite conformational changes in Hsp90. We observed that NB and KU-32 both bind to the C-terminal domain of Hsp90, but surprisingly, KU-32 stimulated the chaperone functions of Hsp90 via allosteric modulation of its N-terminal domain, responsible for the chaperone's ATPase activity. In vitro and in silico studies indicated that upon KU-32 binding, Hsp90 undergoes global structural changes leading to the formation of a "partially closed" intermediate that selectively binds ATP and increases ATPase activity. We also report that KU-32 promotes HeLa cell survival and enhances the refolding of an Hsp90 substrate inside the cell. This discovery explains the effectiveness of KU-32 analogs in the management of neuropathies and may facilitate the design of molecules that promote cell survival by enhancing Hsp90 chaperone function and reducing the load of misfolded proteins in cells.


Asunto(s)
Inhibidores Enzimáticos , Proteínas HSP90 de Choque Térmico , Novobiocina/análogos & derivados , Pliegue de Proteína/efectos de los fármacos , Regulación Alostérica/efectos de los fármacos , Supervivencia Celular/efectos de los fármacos , Inhibidores Enzimáticos/química , Inhibidores Enzimáticos/farmacología , Proteínas HSP90 de Choque Térmico/antagonistas & inhibidores , Proteínas HSP90 de Choque Térmico/química , Proteínas HSP90 de Choque Térmico/metabolismo , Células HeLa , Humanos , Novobiocina/química , Novobiocina/farmacología , Unión Proteica , Dominios Proteicos
5.
J Biol Chem ; 293(34): 13270-13283, 2018 08 24.
Artículo en Inglés | MEDLINE | ID: mdl-29959230

RESUMEN

Most protein folding studies until now focus on single domain or truncated proteins. Although great insights in the folding of such systems has been accumulated, very little is known regarding the proteins containing multiple domains. It has been shown that the high stability of domains, in conjunction with inter-domain interactions, manifests as a frustrated energy landscape, causing complexity in the global folding pathway. However, multidomain proteins despite containing independently foldable, loosely cooperative sections can fold into native states with amazing speed and accuracy. To understand the complexity in mechanism, studies were conducted previously on the multidomain protein malate synthase G (MSG), an enzyme of the glyoxylate pathway with four distinct and adjacent domains. It was shown that the protein refolds to a functionally active intermediate state at a fast rate, which slowly produces the native state. Although experiments decoded the nature of the intermediate, a full description of the folding pathway was not elucidated. In this study, we use a battery of biophysical techniques to examine the protein's folding pathway. By using multiprobe kinetics studies and comparison with the equilibrium behavior of protein against urea, we demonstrate that the unfolded polypeptide undergoes conformational compaction to a misfolded intermediate within milliseconds of refolding. The misfolded product appears to be stabilized under moderate denaturant concentrations. Further folding of the protein produces a stable intermediate, which undergoes partial unfolding-assisted large segmental rearrangements to achieve the native state. This study reveals an evolved folding pathway of the multidomain protein MSG, which involves surpassing the multiple misfolding traps during refolding.


Asunto(s)
Escherichia coli/enzimología , Malato Sintasa/química , Conformación Proteica , Pliegue de Proteína , Replegamiento Proteico , Cristalografía por Rayos X , Cinética , Malato Sintasa/metabolismo , Modelos Moleculares , Desnaturalización Proteica , Termodinámica
6.
Microb Cell Fact ; 18(1): 215, 2019 Dec 17.
Artículo en Inglés | MEDLINE | ID: mdl-31847856

RESUMEN

BACKGROUND: Serratia marcescens, a Gram-negative nosocomial pathogen secretes a 50 kDa multi-domain zinc metalloprotease called serratiopeptidase. Broad substrate specificity of serratiopeptidase makes it suitable for detergent and food processing industries The protein shows potent anti-inflammatory, anti-edemic, analgesic, antibiofilm activity and sold as an individual or fixed-dose enteric-coated tablets combined with other drugs. Although controversial, serratiopeptidase as drug is used in the treatment of chronic sinusitis, carpal tunnel syndrome, sprains, torn ligaments, and postoperative inflammation. Since the native producer of serratiopeptidase is a pathogenic microorganism, the current production methods need to be replaced by alternative approaches. Heterologous expression of serratiopeptidase in E. coli was tried before but not found suitable due to the limited yield, and other expression related issues due to its inherent proteolytic activity such as cytotoxicity, cell death, no expression, minimal expression, or inactive protein accumulation. RESULTS: Recombinant expression of mature form serratiopeptidase in E. coli seems toxic and resulted in the failure of transformation and other expression related issues. Although E. coli C43(DE3) cells, express protein correctly, the yield was compromised severely. Optimization of protein expression process parameters such as nutrient composition, induction point, inducer concentration, post-induction duration, etc., caused significant enhancement in serratiopeptidase production (57.9 ± 0.73% of total cellular protein). Expressed protein formed insoluble, enzymatically inactive inclusion bodies, and gave 40-45 mg/l homogenous (> 98% purity) biologically active and conformationally similar serratiopeptidase to the commercial counterpart upon refolding and purification. CONCLUSION: Expression of mature serratiopeptidase in E. coli C43(DE3) cells eliminated the protein expression associated with toxicity issues. Further optimization of process parameters significantly enhanced the overexpression of protein resulting in the higher yield of pure and functionally active recombinant serratiopeptidase. The biological activity and conformational features of recombinant serratiopeptidase were very similar to the commercially available counterpart suggesting it-a potential biosimilar of therapeutic and industrial relevance.


Asunto(s)
Biosimilares Farmacéuticos/metabolismo , Escherichia coli/enzimología , Péptido Hidrolasas/biosíntesis , Péptido Hidrolasas/química , Péptido Hidrolasas/metabolismo , Conformación Proteica , Proteínas Recombinantes/biosíntesis , Proteínas Recombinantes/química , Proteínas Recombinantes/metabolismo
7.
Int J Med Microbiol ; 308(8): 1000-1008, 2018 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-30190103

RESUMEN

Utility of Mycobacterium indicus pranii (MIP) as a multistage vaccine against mycobacterial infections demands identification of its protective antigens. We explored antigenicity and immunogenicity of a candidate protein MIP_05962 that depicts homology to HSP18 of M. leprae and antigen1 of Mycobacterium tuberculosis. This protein elicited substantial antibody response in immunized mice along with modulation of cellular immune response towards protective Th1 type. Both CD4+ and CD8+ subsets from immunized mice produced hallmark protective cytokines, IFN-γ, TNF-α and IL-2. This protein also enhanced the CD4+ effector memory that could act as first line of defence during infections. These results point to MIP_05962 as a protective antigen that contributes, in conjunction with others, to the protective immunity of this live vaccine candidate.


Asunto(s)
Proteínas Bacterianas/inmunología , ADN Bacteriano/inmunología , Complejo Mycobacterium avium/inmunología , Infección por Mycobacterium avium-intracellulare/inmunología , Células TH1/inmunología , Animales , Proteínas Bacterianas/genética , Citocinas/inmunología , Citocinas/metabolismo , ADN Bacteriano/genética , Humanos , Inmunidad Celular/inmunología , Inmunidad Humoral/inmunología , Inmunización , Ratones , Ratones Endogámicos BALB C , Complejo Mycobacterium avium/genética , Infección por Mycobacterium avium-intracellulare/microbiología , Cultivo Primario de Células , Proteínas Recombinantes/genética , Proteínas Recombinantes/inmunología , Células TH1/metabolismo , Vacunas contra la Tuberculosis/inmunología
8.
Biochim Biophys Acta Proteins Proteom ; 1866(9): 941-951, 2018 09.
Artículo en Inglés | MEDLINE | ID: mdl-29864530

RESUMEN

The isolated apical domain of GroEL consisting of residues 191-345 (known as "minichaperone") binds and assists the folding of a wide variety of client proteins without GroES and ATP, but the mechanism of its action is still unknown. In order to probe into the matter, we have examined minichaperone-mediated folding of a large aggregation prone protein Maltodextrin-glucosidase (MalZ). The key objective was to identify whether MalZ exists free in solution, or remains bound to, or cycling on and off the minichaperone during the refolding process. When GroES was introduced during refolding process, production of the native MalZ was inhibited. We also observed the same findings with a trap mutant of GroEL, which stably captures a predominantly non-native MalZ released from minichaperone during refolding process, but does not release it. Tryptophan and ANS fluorescence measurements indicated that refolded MalZ has the same structure as the native MalZ, but that its structure when bound to minichaperone is different. Surface plasmon resonance measurements provide an estimate for the equilibrium dissociation constant KD for the MalZ-minichaperone complex of 0.21 ±â€¯0.04 µM, which are significantly higher than for most GroEL clients. This showed that minichaperone interacts loosely with MalZ to allow the protein to change its conformation and fold while bound during the refolding process. These observations suggest that the minichaperone works by carrying out repeated cycles of binding aggregation-prone protein MalZ in a relatively compact conformation and in a partially folded but active state, and releasing them to attempt to fold in solution.


Asunto(s)
Chaperonina 60/fisiología , Proteínas de Escherichia coli/metabolismo , Escherichia coli/metabolismo , Glicósido Hidrolasas/metabolismo , Pliegue de Proteína , Chaperonina 60/química , Escherichia coli/genética , Proteínas de Escherichia coli/química , Glicósido Hidrolasas/química , Unión Proteica , Dominios Proteicos , Resonancia por Plasmón de Superficie
9.
Biochim Biophys Acta ; 1864(9): 1138-1151, 2016 09.
Artículo en Inglés | MEDLINE | ID: mdl-27317979

RESUMEN

Maltodextrin glucosidase (MalZ) hydrolyses short malto-oligosaccharides from the reducing end releasing glucose and maltose in Escherichia coli. MalZ is a highly aggregation prone protein and molecular chaperonins GroEL and GroES assist in the folding of this protein to a substantial level. The N-terminal region of this enzyme appears to be a unique domain as seen in sequence comparison studies with other amylases as well as through homology modelling. The sequence and homology model analysis show a probability of disorder in the N-Terminal region of MalZ. The crystal structure of this enzyme has been reported in the present communication. Based on the crystallographic structure, it has been interpreted that the N-terminal region of the enzyme (Met1-Phe131) might be unstructured or flexible. To understand the role of the N-terminal region of MalZ in its enzymatic activity, and overall stability, a truncated version (Ala111-His616) of MalZ was created. The truncated version failed to fold into an active enzyme both in E. coli cytosol and in vitro even with the assistance of chaperonins GroEL and GroES. Furthermore, the refolding effort of N-truncated MalZ in the presence of isolated N-terminal domain didn't succeed. Our studies suggest that while the structural rigidity or orientation of the N-terminal region of the MalZ protein may not be essential for its stability and function, but the said domain is likely to play an important role in the formation of the native structure of the protein when present as an integral part of the protein.


Asunto(s)
Proteínas de Escherichia coli/química , Escherichia coli/química , Glucósidos/química , Glicósido Hidrolasas/química , Secuencia de Aminoácidos , Sitios de Unión , Chaperonina 60/química , Chaperonina 60/genética , Chaperonina 60/metabolismo , Clonación Molecular , Cristalografía por Rayos X , Escherichia coli/enzimología , Escherichia coli/genética , Proteínas de Escherichia coli/genética , Proteínas de Escherichia coli/metabolismo , Expresión Génica , Glucósidos/metabolismo , Glicósido Hidrolasas/genética , Glicósido Hidrolasas/metabolismo , Modelos Moleculares , Agregado de Proteínas , Unión Proteica , Conformación Proteica en Hélice alfa , Conformación Proteica en Lámina beta , Pliegue de Proteína , Dominios y Motivos de Interacción de Proteínas , Proteínas Recombinantes/química , Proteínas Recombinantes/genética , Proteínas Recombinantes/metabolismo , Alineación de Secuencia , Homología Estructural de Proteína , Especificidad por Sustrato
10.
Microb Cell Fact ; 16(1): 173, 2017 Oct 05.
Artículo en Inglés | MEDLINE | ID: mdl-28982367

RESUMEN

BACKGROUND: Human serum albumin (HSA)-one of the most demanded therapeutic proteins with immense biotechnological applications-is a large multidomain protein containing 17 disulfide bonds. The current source of HSA is human blood plasma which is a limited and unsafe source. Thus, there exists an indispensable need to promote non-animal derived recombinant HSA (rHSA) production. Escherichia coli is one of the most convenient hosts which had contributed to the production of more than 30% of the FDA approved recombinant pharmaceuticals. It grows rapidly and reaches high cell density using inexpensive and simple subst-rates. E. coli derived recombinant products have more economic potential as fermentation processes are cheaper compared to the other expression hosts. The major bottleneck in exploiting E. coli as a host for a disulfide-rich multidomain protein is the formation of aggregates of overexpressed protein. The majority of the expressed HSA forms inclusion bodies (more than 90% of the total expressed rHSA) in the E. coli cytosol. Recovery of functional rHSA from inclusion bodies is not preferred because it is difficult to obtain a large multidomain disulfide bond rich protein like rHSA in its functional native form. Purification is tedious, time-consuming, laborious and expensive. Because of such limitations, the E. coli host system was neglected for rHSA production for the past few decades despite its numerous advantages. RESULTS: In the present work, we have exploited the capabilities of E. coli as a host for the enhanced functional production of rHSA (~ 60% of the total expressed rHSA in the soluble fraction). Parameters like intracellular environment, temperature, induction type, duration of induction, cell lysis conditions etc. which play an important role in enhancing the level of production of the desired protein in its native form in vivo have been optimized. We have studied the effect of assistance of different types of exogenously employed chaperone systems on the functional expression of rHSA in the E. coli host system. Different aspects of cell growth parameters during the production of rHSA in presence and absence of molecular chaperones in E. coli have also been studied. CONCLUSION: In the present case, we have filled in the gap in the literature by exploiting the E. coli host system, which is fast-growing and scalable at the low cost of fermentation, as a microbial factory for the enhancement of functional production of rHSA, a crucial protein for therapeutic and biotechnological applications.


Asunto(s)
Escherichia coli/genética , Escherichia coli/metabolismo , Albúmina Sérica Humana/biosíntesis , Biotecnología/métodos , Escherichia coli/crecimiento & desarrollo , Humanos , Cuerpos de Inclusión/metabolismo , Ingeniería Metabólica , Chaperonas Moleculares/metabolismo , Proteínas Recombinantes/biosíntesis , Proteínas Recombinantes/química , Proteínas Recombinantes/metabolismo , Albúmina Sérica Humana/química , Albúmina Sérica Humana/metabolismo
11.
Proc Natl Acad Sci U S A ; 110(51): 20467-72, 2013 Dec 17.
Artículo en Inglés | MEDLINE | ID: mdl-24282299

RESUMEN

Resistin, a cysteine-rich adipocytokine, proposed as a link between obesity and diabetes in mice, was shown as a proinflammatory molecule in humans. We earlier reported that human resistin (hRes), a trimer, was resistant to heat and urea denaturation, existed in an oligomeric polydispersed state, and showed a concentration-dependent conformational change. These properties and an intimate correlation of hRes expression with cellular stress prompted us to investigate hRes as a possible chaperone. Here, we show that recombinant human resistin was able to protect the heat-labile enzymes citrate synthase and Nde1 from thermal aggregation and inactivation and was able to refold and restore their enzymatic activities after heat/guanidinium chloride denaturation. Furthermore, recombinant human resistin could bind misfolded proteins only. Molecular dynamics-based association-dissociation kinetics of hRes subunits pointed to resistin being a molecular chaperone. Bis-ANS, which blocks surface hydrophobicity, abrogated the chaperone activity of hRes, establishing the importance of surface hydrophobicity for chaperone activity. Replacement of Phe49 with Tyr (F49YhRes), a critical residue within the hydrophobic patch of hRes, although it could prevent thermal aggregation of citrate synthase and Nde1, was unable to refold and restore their activities. Treatment of U937 cells with tunicamycin/thapsigargin resulted in reduced hRes secretion and concomitant localization in the endoplasmic reticulum. Escherichia coli transformants expressing hRes could be rescued from thermal stress, pointing to hRes's chaperone-like function in vivo. HeLa cells transfected with hRes showed protection from thapsigargin-induced apoptosis. In conclusion, hRes, an inflammatory protein, additionally exhibited chaperone-like properties, suggesting a possible link between inflammation and cellular stress.


Asunto(s)
Citocinas/metabolismo , Respuesta al Choque Térmico/fisiología , Mediadores de Inflamación/metabolismo , Proteínas Asociadas a Microtúbulos/metabolismo , Chaperonas Moleculares/metabolismo , Resistina/metabolismo , Animales , Antibacterianos/farmacología , Citocinas/genética , Inhibidores Enzimáticos/farmacología , Células HeLa , Respuesta al Choque Térmico/efectos de los fármacos , Humanos , Inflamación/genética , Inflamación/metabolismo , Ratones , Proteínas Asociadas a Microtúbulos/genética , Chaperonas Moleculares/genética , Resistina/genética , Tapsigargina/farmacología , Tunicamicina/farmacología , Células U937
12.
J Biol Chem ; 289(1): 286-98, 2014 Jan 03.
Artículo en Inglés | MEDLINE | ID: mdl-24247249

RESUMEN

Despite a vast amount information on the interplay of GroEL, GroES, and ATP in chaperone-assisted folding, the molecular details on the conformational dynamics of folding polypeptide during its GroEL/GroES-assisted folding cycle is quite limited. Practically no such studies have been reported to date on large proteins, which often have difficulty folding in vitro. The effect of the GroEL/GroES chaperonin system on the folding pathway of an 82-kDa slow folding protein, malate synthase G (MSG), was investigated. GroEL bound to the burst phase intermediate of MSG and accelerated the slowest kinetic phase associated with the formation of native topology in the spontaneous folding pathway. GroEL slowly induced conformational changes on the bound burst phase intermediate, which was then transformed into a more folding-compatible form. Subsequent addition of ATP or GroES/ATP to the GroEL-MSG complex led to the formation of the native state via a compact intermediate with the rate several times faster than that of spontaneous refolding. The presence of GroES doubled the ATP-dependent reactivation rate of bound MSG by preventing multiple cycles of its GroEL binding and release. Because GroES bound to the trans side of GroEL-MSG complex, it may be anticipated that confinement of the substrate underneath the co-chaperone is not required for accelerating the rate in the assisted folding pathway. The potential role of GroEL/GroES in assisted folding is most likely to modulate the conformation of MSG intermediates that can fold faster and thereby eliminate the possibility of partial aggregation caused by the slow folding intermediates during its spontaneous refolding pathway.


Asunto(s)
Chaperonina 10/metabolismo , Chaperonina 60/metabolismo , Proteínas de Escherichia coli/metabolismo , Escherichia coli/metabolismo , Malato Sintasa/metabolismo , Replegamiento Proteico , Adenosina Trifosfato/química , Adenosina Trifosfato/genética , Adenosina Trifosfato/metabolismo , Chaperonina 10/química , Chaperonina 10/genética , Chaperonina 60/química , Chaperonina 60/genética , Escherichia coli/química , Escherichia coli/genética , Proteínas de Escherichia coli/química , Proteínas de Escherichia coli/genética , Malato Sintasa/química , Malato Sintasa/genética , Unión Proteica , Estructura Terciaria de Proteína
15.
FEBS J ; 291(9): 1958-1973, 2024 May.
Artículo en Inglés | MEDLINE | ID: mdl-38700222

RESUMEN

Serratia marcescens is an emerging health-threatening, gram-negative opportunistic pathogen associated with a wide variety of localized and life-threatening systemic infections. One of the most crucial virulence factors produced by S. marcescens is serratiopeptidase, a 50.2-kDa repeats-in-toxin (RTX) family broad-specificity zinc metalloprotease. RTX family proteins are functionally diverse exoproteins of gram-negative bacteria that exhibit calcium-dependent structural dynamicity and are secreted through a common type-1 secretion system (T1SS) machinery. To evaluate the impact of various divalent ligands on the folding and maturation of serratiopeptidase zymogen, the protein was purified and a series of structural and functional investigations were undertaken. The results indicate that calcium binding to the C-terminal RTX domain acts as a folding switch, triggering a disordered-to-ordered transition in the enzyme's conformation. Further, the auto-processing of the 16-amino acid N-terminal pro-peptide results in the maturation of the enzyme. The binding of calcium ions to serratiopeptidase causes a highly cooperative conformational transition in its structure, which is essential for the enzyme's activation and maturation. This conformational change is accompanied by an increase in solubility and enzymatic activity. For efficient secretion and to minimize intracellular toxicity, the enzyme needs to be in an unfolded extended form. The calcium-rich extracellular environment favors the folding and processing of zymogen into mature serratiopeptidase, i.e., the holo-form required by S. marcescens to establish infections and survive in different environmental niches.


Asunto(s)
Calcio , Precursores Enzimáticos , Péptido Hidrolasas , Pliegue de Proteína , Serratia marcescens , Calcio/metabolismo , Serratia marcescens/enzimología , Serratia marcescens/genética , Precursores Enzimáticos/metabolismo , Precursores Enzimáticos/química , Precursores Enzimáticos/genética , Metaloendopeptidasas/química , Metaloendopeptidasas/metabolismo , Metaloendopeptidasas/genética , Modelos Moleculares , Conformación Proteica , Proteínas Bacterianas/química , Proteínas Bacterianas/metabolismo , Proteínas Bacterianas/genética , Unión Proteica
16.
ACS Biomater Sci Eng ; 10(8): 4812-4822, 2024 Aug 12.
Artículo en Inglés | MEDLINE | ID: mdl-38976823

RESUMEN

RNA bacteriophage MS2-derived virus-like particles (VLPs) have been widely used in biomedical research as model systems to study virus assembly, structure-function relationships, vaccine development, and drug delivery. Considering the diverse utility of these VLPs, a systemic engineering approach has been utilized to generate smaller particles with optimal serum stability and tissue penetrance. Additionally, it is crucial to demonstrate the overall stability of these mini MS2 VLPs, ensuring cargo protection until they reach their target cell/organ. However, no detailed analysis of the thermal stability and heat-induced disassembly of MS2 VLPs has yet been attempted. In this work, we investigated the thermal stability of both wild-type (WT) MS2 VLP and its "mini" variant containing S37P mutation (mini MS2 VLP). The mini MS2 VLP exhibits a higher capsid melting temperature (Tm) when compared to its WT MS2 VLP counterpart, possibly attributed to its smaller interdimer angle. Our study presents that the thermal unfolding of MS2 VLPs follows a sequential process involving particle destabilization, nucleic acid exposure/melting, and disassembly of VLP. This observation underscores the disruption of cooperative intersubunit interactions and protein-nucleic acid interactions, shedding light on the mechanism of heat-induced VLP disassembly.


Asunto(s)
Levivirus , Levivirus/genética , Levivirus/química , Levivirus/metabolismo , Proteínas de la Cápside/química , Proteínas de la Cápside/metabolismo , Proteínas de la Cápside/genética , Temperatura , Mutación , Calor , Virión/metabolismo , Virión/química , Virión/genética , Cápside/metabolismo , Cápside/química
17.
Biochemistry ; 52(26): 4517-30, 2013 Jul 02.
Artículo en Inglés | MEDLINE | ID: mdl-23718231

RESUMEN

Despite their prevalence in biological systems, information about the folding pathways of large and multidomain proteins is meager, as they often unfold irreversibly under in vitro conditions which make their folding studies difficult or even impossible. The folding mechanism of a large (82 kDa) and multidomain protein Malate synthase G (MSG) has been demonstrated in the present study using intrinsic tryptophan fluorescence, enzymatic activity, and extrinsic fluorophore ANS as probes for monitoring the refolding process. Refolding of MSG is found to occur in three kinetic phases. Denatured MSG forms a collapsed state in the burst phase of refolding, which then gives rise to an active intermediate having the same tryptophan fluorescence and enzymatic activity as native MSG in the slow phase. Native topology of MSG is formed from the active intermediate in the very slow phase of refolding which is silent to tryptophan fluorescence change and is susceptible to aggregation at higher protein concentrations. Dependence of rates of very slow phase on GdnHCl concentration suggests that it is not solely a cis/trans proline isomerization limited process but might involve an additional folding event of the domains, not forming the active site of the protein. In light of the above findings, the appearance of a functional intermediate during refolding of MSG was predicted to be an instance of weak interdomain cooperativity. This work has significant implications in the characterization of the refolding intermediates of multidomain proteins in general and MSG in particular, where weak interdomain cooperativity might contribute toward generation of a functional intermediate during its refolding.


Asunto(s)
Escherichia coli/enzimología , Malato Sintasa/química , Conformación Proteica , Pliegue de Proteína , Dicroismo Circular , Clonación Molecular , Cristalografía por Rayos X , Fluorescencia , Cinética , Prolina/química , Desnaturalización Proteica , Estructura Terciaria de Proteína , Termodinámica
18.
J Biomol Struct Dyn ; : 1-9, 2023 Aug 14.
Artículo en Inglés | MEDLINE | ID: mdl-37578048

RESUMEN

Trichostatin A (TSA), a potential radiomitigator in pre-clinical models, inhibits the class I and II mammalian histone deacetylase (HDAC) enzyme family preferentially. In the current study, the ADME assessment of TSA was explored in terms of its binding affinity for serum protein via spectroscopic and molecular docking techniques. Fluorescence spectroscopy was used to examine changes in the protein microenvironment, and affinity was quantified in terms of binding constant and stoichiometry. Post binding conformational changes were observed using circular dichroism (CD) and UV-Visible spectroscopy. Specific binding was visualized using molecular docking to support experimental studies. UV-vis spectra demonstrated a blue shift in the interaction of TSA to BSA. The calculated binding constants ranged from 3.10 to 0.78 x 10 5(M-1) and quenching constants from 2.75 to 2.15 x 104 (l mol-1), indicating TSA has a strong binding affinity for BSA. Based on the FRET theory, the distance between BSA (donor) and TSA (acceptor) was calculated to be 2.83 nm. The Stern-Volmer plot revealed (Ksv) static quenching. Thermodynamic parameters were calculated, and a negative ΔG value showed that the interaction is spontaneous. The CD spectra analysis further revealed a change in the protein's secondary structure, indicating TSA-BSA interaction. The molecular docking studies also indicated strong binding affinity of TSA with BSA. The results indicate that good bio-availability of TSA is possible because of the spontaneous and strong binding affinity with BSA.Communicated by Ramaswamy H. Sarma.

19.
J Biomol Struct Dyn ; : 1-18, 2023 Aug 11.
Artículo en Inglés | MEDLINE | ID: mdl-37565730

RESUMEN

Breast cancer is one of the most prevalent and malignant cancers in women. Most breast cancer patients show overexpression of the HER2 protein. The current study focused on identifying potent inhibitors of HER2 using a structure-based drug design approach. Prefiltered compounds from the Drugbank and the ZINC database were docked on HER2 protein using the FlexX docking tool of LeadIT. The docking study identified the 12 best molecules that interacted strongly with the active site of HER2 and also fulfilled the ADMET parameters. The complexes of these compounds with HER2 were further subjected to molecular dynamics simulation using GROMACS 2021.4, followed by the end-state MMGBSA binding energy calculations. The RMSD analysis was conducted to study the conformational changes, which revealed stability throughout the 100 ns simulation period. The local flexibility and dynamics of the simulated ligand-protein complexes were studied using RMSF analysis. The values of the radius of gyration were computed to analyze the compactness of HER2. The MMGBSA analysis provided insights into the energetic aspects of the system. The compound DB15187 emerged as the most potent candidate, showing MMGBSA-computed binding energy of -63.60 ± 3.39 kcal/mol. The study could help develop targeted therapies for HER2-positive breast cancer.Communicated by Ramaswamy H. Sarma.

20.
BMC Genomics ; 13 Suppl 7: S22, 2012.
Artículo en Inglés | MEDLINE | ID: mdl-23281895

RESUMEN

BACKGROUND: Molecular chaperones appear to have been evolved to facilitate protein folding in the cell through entrapment of folding intermediates on the interior of a large cavity formed between GroEL and its co-chaperonin GroES. They bind newly synthesized or non-native polypeptides through hydrophobic interactions and prevent their aggregation. Some proteins do not interact with GroEL, hence even though they are aggregation prone, cannot be assisted by GroEL for their folding. RESULTS: In this study, we have attempted to engineer these non-substrate proteins to convert them as the substrate for GroEL, without compromising on their function. We have used a computational biology approach to generate mutants of the selected proteins by selectively mutating residues in the hydrophobic patch, similar to GroES mobile loop region that are responsible for interaction with GroEL, and compared with the wild counterparts for calculation of their instability and aggregation propensities. The energies of the newly designed mutants were computed through molecular dynamics simulations. We observed increased aggregation propensity of some of the mutants formed after replacing charged amino acid residues with hydrophobic ones in the well defined hydrophobic patch, raising the possibility of their binding ability to GroEL. CONCLUSIONS: The newly generated mutants may provide potential substrates for Chaperonin GroEL, which can be experimentally generated and tested for their tendency of aggregation, interactions with GroEL and the possibility of chaperone-assisted folding to produce functional proteins.


Asunto(s)
Chaperonina 60/metabolismo , Ingeniería de Proteínas , Proteínas Recombinantes/metabolismo , Secuencia de Aminoácidos , Chaperonina 60/química , Biología Computacional , Bases de Datos de Proteínas , Interacciones Hidrofóbicas e Hidrofílicas , Simulación de Dinámica Molecular , Proteínas Recombinantes/genética , Especificidad por Sustrato
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA