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










Base de datos
Intervalo de año de publicación
1.
bioRxiv ; 2023 Jul 03.
Artículo en Inglés | MEDLINE | ID: mdl-37461525

RESUMEN

Loss of proteostasis is a hallmark of aging and Alzheimer disease (AD). Here, we identify ß-hydroxybutyrate (ßHB), a ketone body, as a regulator of protein solubility in the aging brain. ßHB is a small molecule metabolite which primarily provides an oxidative substrate for ATP during hypoglycemic conditions, and also regulates other cellular processes through covalent and noncovalent protein interactions. We demonstrate ßHB-induced protein insolubility across in vitro, ex vivo, and in vivo mouse systems. This activity is shared by select structurally similar metabolites, is not dependent on covalent protein modification, pH, or solute load, and is observable in mouse brain in vivo after delivery of a ketone ester. Furthermore, this phenotype is selective for pathological proteins such as amyloid-ß, and exogenous ßHB ameliorates pathology in nematode models of amyloid-ß aggregation toxicity. We have generated a comprehensive atlas of the ßHB-induced protein insolublome ex vivo and in vivo using mass spectrometry proteomics, and have identified common protein domains within ßHB target sequences. Finally, we show enrichment of neurodegeneration-related proteins among ßHB targets and the clearance of these targets from mouse brain, likely via ßHB-induced autophagy. Overall, these data indicate a new metabolically regulated mechanism of proteostasis relevant to aging and AD.

2.
Age (Dordr) ; 36(3): 9597, 2014 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-24254744

RESUMEN

Bivalve mollusks have several unique traits, including some species with exceptionally long lives, others with very short lives, and the ability to determine the age of any individual from growth rings in the shell. Exceptionally long-lived species are seldom studied yet have the potential to be particularly informative with respect to senescence-resistance mechanisms. To this end, we employed a range of marine bivalve mollusk species, with lifespans ranging from under a decade to over 500 years, in a comparative study to investigate the hypothesis that long life requires superior proteome stability. This experimental system provides a unique opportunity to study closely related organisms with vastly disparate longevities, including the longest lived animal, Arctica islandica.Specifically, we investigated relative ability to protect protein structure and function, both basally and under various stressors in our range of species. We found a consistent relationship between species longevity, resistance to protein unfolding, and maintenance of endogenous enzyme (creatine kinase) activity. Remarkably, our longest-lived species, Arctica islandica (maximum longevity >500 years), had no increase in global proteome unfolding in response to several stressors. Additionally, the global proteome of shorter-lived species exhibited less resistance to temperature-induced protein aggregation than longer-lived species. A reporter assay, in which the same protein's aggregation properties was assessed in lysates from each study species, suggests that some endogenous feature in the cells of long-lived species, perhaps small molecular chaperones, was at least partially responsible for their enhanced proteome stability. This study reinforces the relationship between proteostasis and longevity through assessment of unfolding, function, and aggregation in species ranging in longevity from less than a decade to more than five centuries.


Asunto(s)
Envejecimiento/fisiología , Bivalvos/fisiología , Longevidad/fisiología , Proteoma/metabolismo , Animales
3.
Neurology ; 68(5): 326-37, 2007 Jan 30.
Artículo en Inglés | MEDLINE | ID: mdl-17261678

RESUMEN

OBJECTIVE: To estimate the current incidence and prevalence in the United States of 12 neurologic disorders. METHODS: We summarize the strongest evidence available, using data from the United States or from other developed countries when US data were insufficient. RESULTS: For some disorders, prevalence is a better descriptor of impact; for others, incidence is preferable. Per 1,000 children, estimated prevalence was 5.8 for autism spectrum disorder and 2.4 for cerebral palsy; for Tourette syndrome, the data were insufficient. In the general population, per 1,000, the 1-year prevalence for migraine was 121, 7.1 for epilepsy, and 0.9 for multiple sclerosis. Among the elderly, the prevalence of Alzheimer disease was 67 and that of Parkinson disease was 9.5. For diseases best described by annual incidence per 100,000, the rate for stroke was 183, 101 for major traumatic brain injury, 4.5 for spinal cord injury, and 1.6 for ALS. CONCLUSIONS: Using the best available data, our survey of a limited number of disorders shows that the burden of neurologic illness affects many millions of people in the United States.


Asunto(s)
Enfermedades del Sistema Nervioso/epidemiología , Modelos de Riesgos Proporcionales , Medición de Riesgo/métodos , Sesgo , Interpretación Estadística de Datos , Países Desarrollados/estadística & datos numéricos , Humanos , Incidencia , Prevalencia , Reproducibilidad de los Resultados , Factores de Riesgo , Tamaño de la Muestra , Estados Unidos/epidemiología
4.
Biochemistry ; 40(30): 8834-41, 2001 Jul 31.
Artículo en Inglés | MEDLINE | ID: mdl-11467944

RESUMEN

Cysteine residues in tubulin are actively involved in regulating ligand interactions and microtubule formation both in vivo and in vitro. These cysteine residues are sensitive reporters in determining the conformation of tubulin. Although some of the cysteines are critical in modulating drug binding and microtubule assembly, it is not clear how many of these normally exist as disulfides. The controversy regarding the disulfide bonds led us to develop a disulfide detection assay to reexamine the presence of the disulfide linkages in purified alphabeta tubulin and explore their possible biological functions in vitro. The accessible cysteine residues in alphabeta tubulin were alkylated with an excess of iodoacetamide to prevent artifactual generation of disulfide linkages in tubulin. After removal of excess iodoacetamide, tubulin was unfolded in 8 M urea. Half of the unfolded tubulin was treated with dithiothreitol to reduce any disulfide bonds present. The aliquots were then treated with iodo[(14)C]acetamide and the incorporation of radioactivity was measured. We also used the same approach to detect the disulfide linkages in the tubulin in a whole-cell extract. We found in both cases that the samples which were not treated with dithiothreitol had little or no incorporation of iodo[(14)C]acetamide, while the others that were treated with dithiothreitol had significant amounts of (14)C incorporation into tubulin. Moreover, the reduction of the disulfide linkages in tubulin resulted in inhibition of microtubule assembly (29-54%) and markedly affected refolding of the tubulin from both an intermediate and a completely unfolded state. All these data therefore suggest that tubulin has intrachain disulfide bonds in the alpha- and beta-subunits and that these disulfides assist in correct refolding of tubulin from the intermediate unfolded state or help to recover the hydrophobic domains from the completely unfolded state. These disulfides also regulate microtubule assembly and the stability of tubulin in vitro. Our results suggest that tubulin disulfides may play a role in tubulin folding and that thiol-disulfide exchange in tubulin could be a key regulator in microtubule assembly and dynamics of tubulin in vivo.


Asunto(s)
Química Encefálica , Disulfuros/química , Microtúbulos/metabolismo , Pliegue de Proteína , Tubulina (Proteína)/química , Sulfato de Amonio , Animales , Química Encefálica/efectos de los fármacos , Bovinos , Colchicina/metabolismo , Cisteína/metabolismo , Disulfuros/aislamiento & purificación , Ditiotreitol/metabolismo , Ditiotreitol/farmacología , Microtúbulos/química , Unión Proteica , Conformación Proteica , Desnaturalización Proteica , Sustancias Reductoras/metabolismo , Sustancias Reductoras/farmacología , Espectrometría de Fluorescencia , Triptófano/metabolismo , Tubulina (Proteína)/aislamiento & purificación , Tubulina (Proteína)/metabolismo , Urea
5.
J Indian Med Assoc ; 99(8): 445-7, 2001 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-11881860

RESUMEN

Standard approach towards treatment of obstructive azoospermia whether congenital or acquired is mostly microsurgical epididymal sperm aspiration (MESA) and to some extent percutaneous epididymal sperm aspiration (PESA) both being followed by intracytoplasmic sperm injection (ICSI). However in the present series for sperm retrieval in case of obstructive azoospermia the author conducted a series of 55 cases of PESA along with only 4 cases of MESA. The epididymal sperm aspiration (ESA) was followed by intrauterine insemination (IUI) (whenever required quantity of motile sperm was collected) in place of ICSI with full consent of the couples. With the help of this ESA-IUI programme it was possible to achieve reasonable success rate viz, 20% pregnancy rate (PR) per cycle for PESA-IUI and 14.28% for MESA-IUI.


Asunto(s)
Infertilidad Masculina/terapia , Inseminación Artificial/métodos , Oligospermia/terapia , Embarazo/estadística & datos numéricos , Inyecciones de Esperma Intracitoplasmáticas/métodos , Adulto , Epidídimo/citología , Femenino , Estudios de Seguimiento , Humanos , Masculino , Microcirugia/métodos , Oligospermia/etiología , Estudios Retrospectivos , Sensibilidad y Especificidad , Resultado del Tratamiento
6.
J Mol Biol ; 303(5): 679-92, 2000 Nov 10.
Artículo en Inglés | MEDLINE | ID: mdl-11061968

RESUMEN

Tubulin, the major structural component of the microtubules, participates actively in mitotic spindle formation and chromosomal organization during cell division. Tubulin is the major target for a variety of anti-mitotic drugs. Some of the drugs, such as Vinca alkaloids and taxol, are routinely used for cancer chemotherapy. It is unfortunate that our knowledge of the binding sites on tubulin of these drugs is limited because of lack of a useful and appropriate tool. The photoaffinity labeling approach is the major technique available at present to detect the binding sites of drugs on tubulin. This method, however, has several limitations. First, only part of the binding site can be identified, namely, the residues which react with the photoaffinity label. Second, there are regions of tubulin which are not at the binding site but are affected by the binding of the drug; these regions can not be detected by the photoaffinity labeling approach. The third, and perhaps most serious, limitation is that the traditional approach can detect areas which have nothing to do with the binding of the ligand but which are within a certain distance of the binding site, that distance being less than the length of the photoreactive moiety attached to the ligand. There has been a great deal of controversy on the localization of the binding site of colchicine on tubulin, with some reports suggesting that the binding site is on alpha and some supporting a binding site on beta. Colchicine also has significant effects on tubulin conformation, but the regions which are affected have not been identified. We have attempted here to address these questions by a novel "footprinting" method by which the drug-binding sites and as well as the domain of tubulin affected by drug-induced conformational changes could be determined. Here, we report for the first time that the interaction of the B-ring of colchicine with the alpha-subunit affects a domain of tubulin which appears to be far from its binding site. This domain includes the cysteine residues at positions 295, 305, 315 and 316 on alpha-tubulin; these residues are located well away from the alpha/beta interface where colchicine appears to bind. This is correlated with the stabilizing effect of colchicine on the tubulin molecule. Furthermore, we also found that the B-ring of colchicine plays a major role in the stability of tubulin while the A and the C-rings have little effect on it. Our results therefore, support a model whereby colchicine binds at the alpha/beta interface of tubulin with the B-ring on the alpha-subunit and the A and the C-rings on the beta-subunit.


Asunto(s)
Colchicina/química , Colchicina/metabolismo , Huella de Proteína , Estilbenos , Tropolona/análogos & derivados , Tubulina (Proteína)/química , Tubulina (Proteína)/metabolismo , Secuencia de Aminoácidos , Animales , Bibencilos/química , Bibencilos/metabolismo , Bibencilos/farmacología , Sitios de Unión , Bovinos , Cromatografía Líquida de Alta Presión , Colchicina/farmacología , Fluorescencia , Modelos Moleculares , Datos de Secuencia Molecular , Fragmentos de Péptidos/química , Fragmentos de Péptidos/metabolismo , Podofilotoxina/química , Podofilotoxina/metabolismo , Podofilotoxina/farmacología , Conformación Proteica/efectos de los fármacos , Tropolona/química , Tropolona/metabolismo , Tropolona/farmacología , Triptófano/metabolismo , Urea/farmacología
7.
Neurochem Res ; 25(1): 19-25, 2000 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-10685600

RESUMEN

Pyridostigmine bromide (PB), an inhibitor of acetylcholinesterase, has been used as a prophylactic for nerve gas poisoning. N,N'-diethyl-m-toluamide (DEET) is the active ingredient in most insect repellents and is thought to interact synergistically with PB. Since PB can inhibit the binding of organophosphates to tubulin and since organophosphates inhibit microtubule assembly, we decided to examine the effects of PB and DEET on microtubule assembly as well as their interactions with tubulin, the subunit protein of microtubules. We found that PB binds to tubulin with an apparent Kd of about 60 microM. PB also inhibits microtubule assembly in vitro, although at higher concentrations PB induces formation of tubulin aggregates of high absorbance. Like PB, DEET is a weak inhibitor of microtubule assembly and also induces formation of tubulin aggregates. Many tubulin ligands stabilize the conformation of tubulin as measured by exposure of sulfhydryl groups and hydrophobic areas and stabilization of colchicine binding. PB appears to have very little effect on tubulin conformation, and DEET appears to have no effect. Neither compound interferes with colchicine binding to tubulin. Our results raise the possibility that PB and DEET may exert some of their effects in vivo by interfering with microtubule assembly or function, although high intracellular levels of these compounds would be required.


Asunto(s)
Química Encefálica , Inhibidores de la Colinesterasa/farmacología , DEET/farmacología , Repelentes de Insectos/farmacología , Bromuro de Piridostigmina/farmacología , Tubulina (Proteína)/química , Animales , Bovinos , Inhibidores de la Colinesterasa/metabolismo , Colchicina/metabolismo , Cinética , Microtúbulos/efectos de los fármacos , Conformación Proteica/efectos de los fármacos , Bromuro de Piridostigmina/metabolismo , Tritio , Tubulina (Proteína)/metabolismo
8.
J Biol Chem ; 274(34): 24378-82, 1999 Aug 20.
Artículo en Inglés | MEDLINE | ID: mdl-10446217

RESUMEN

FHIT (fragile histidine triad) is a candidate human tumor suppressor gene located at chromosome 3p14.2, a location that encompasses the FRA3B chromosomal fragile site. Aberrant transcripts have been detected in a variety of primary tumors, and homozygous deletions in the FHIT locus have been detected in different tumor cell lines. The gene product Fhit in vitro possesses the ability to hydrolyze diadenosine 5',5"'-P(1),P(3)-triphosphate (Ap(3)A). The mechanism of action of Fhit as a tumor suppressor is unknown. Because the tubulin-microtubule system plays an important role in cell division and cell proliferation, we investigated the interaction between wild-type Fhit or mutant Fhit (H96N) and tubulin in vitro. The mutant form of Fhit (H96N) lacks Ap(3)A hydrolase activity but retains tumor suppressor activity. We found that both wild-type and mutated forms of Fhit bind to tubulin strongly and specifically with K(d) values of 1.4 and 2.1 microM, respectively. Neither wild-type nor mutant Fhit cause nucleation or formation of microtubules, but in the presence of microtubule-associated proteins, both wild-type and mutant Fhit promote assembly to a greater extent than do microtubule-associated proteins alone, and the microtubules formed appear normal by electron microscopy. Our results suggest the possibility that Fhit may exert its tumor suppressor activity by interacting with microtubules and also indicate that the interaction between Fhit and tubulin is not related to the Ap(3)A hydrolase activity of Fhit.


Asunto(s)
Genes Supresores de Tumor , Proteínas de Neoplasias , Proteínas/metabolismo , Tubulina (Proteína)/metabolismo , Ácido Anhídrido Hidrolasas/metabolismo , Proteína de la Poliposis Adenomatosa del Colon , Animales , Bovinos , Proteínas del Citoesqueleto/fisiología , Microscopía Electrónica , Proteínas Asociadas a Microtúbulos/fisiología , Microtúbulos/química , Microtúbulos/fisiología , Microtúbulos/ultraestructura , Proteínas/genética , Proteínas tau/fisiología
9.
Exp Hematol ; 27(7): 1194-203, 1999 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-10390195

RESUMEN

Fas-mediated apoptosis is one of the immune effector pathways leading to the elimination of virus infected cells. In vivo, apoptotic signals are delivered to virus infected cells by Fas-L and other cytokines secreted by specific T lymphocytes. Cellular immune response appears to be essential in prevention of human cytomegalovirus (HCMV) disease. We have hypothesized that HCMV infection might directly or indirectly result in upregulation of Fas receptor and in the presence of Fas ligand, lead to apoptosis of infected cells. We show that infection of human fibroblasts with HCMV is associated with upmodulation of Fas-R process that could be further potentiated by interferon (IFN-gamma). Using DNA agarose gel electrophoresis, terminal dideoxy transferase reaction, and annexin assay, we demonstrated that in a productive HCMV infection of human fibroblasts, loss of cell viability was not only due to virus-mediated cell lysis but also to due to apoptosis. IFN-gamma induced relative HCMV resistance and prevented loss in cell viability. In contrast, anti-Fas monoclonal antibody CH11, serving as Fas agonist, resulted in an accelerated loss in viability of infected cells. IFN-gamma in combination with CH11 further increased the rate of apoptosis and compared to cultures with CH11 only, this effect was not restricted to only infected cells. While IFN-gamma did not affect the number of cells expressing immediate early antigen, it markedly reduced structural protein expression. IFN-gamma in combination with CH11, decreased the expression of HCMV matrix protein pp65, reduced the amount of HCMV DNA and infectious virus produced. Our results are consistent with the theory that cells infected with HCMV can be eliminated by immune effector cells via Fas-mediated apoptosis. IFN-gamma, in addition to its intrinsic antiviral activity, primes HCMV infected cells to the action of Fas ligand and Fas-mediated apoptosis.


Asunto(s)
Apoptosis/efectos de los fármacos , Citomegalovirus/fisiología , Interferón gamma/farmacología , Glicoproteínas de Membrana/fisiología , Receptor fas/fisiología , Anticuerpos Monoclonales/farmacología , Células Cultivadas , Citomegalovirus/efectos de los fármacos , Citomegalovirus/aislamiento & purificación , Infecciones por Citomegalovirus/tratamiento farmacológico , Infecciones por Citomegalovirus/inmunología , Infecciones por Citomegalovirus/virología , Fragmentación del ADN , Proteína Ligando Fas , Humanos , Inmunidad Celular , Linfocinas/metabolismo , Proteínas Recombinantes , Linfocitos T/metabolismo , Ensayo de Placa Viral , Proteínas Virales/análisis , Replicación Viral/efectos de los fármacos
10.
J Protein Chem ; 17(7): 663-8, 1998 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-9853681

RESUMEN

Tubulin, the subunit protein of microtubules, undergoes a time-dependent loss of functional properties known as decay. We have previously shown that the drug 2-(4-fluorophenyl)- -(2-chloro-3,5-dimethoxyphenyl)-3-methyl-6-phenyl-4(1H)-pyridinone (IKP104) accelerates decay, but that in the presence of colchicine, IKP104 becomes a stabilizer of tubulin. To see if this is due to conformational effects specific to colchicine or simply to occupancy at the colchicine site, we examined the effects of nocodazole and podophyllotoxin, two well-known competitive inhibitors of colchicine for binding to tubulin, on IKP104's acceleration of decay. We found that podophyllotoxin abolished IKP104's accelerating effect and, like colchicine, turned it into a stabilizer of tubulin. Nocodazole's effects were similar to those of podophyllotoxin and colchicine, in that it abolished IKP104-induced enhancement of decay; however, in the presence of nocodazole, IKP104 caused little or no stabilization of tubulin. Since colchicine, nocodazole, and podophyllotoxin have very different interactions with tubulin, but all inhibit the IKP104-induced enhancement of decay, our findings suggest that this inhibition arises from occupancy of the colchicine site rather than from a direct conformational effect of these two drugs.


Asunto(s)
Antineoplásicos/farmacología , Nocodazol/farmacología , Podofilotoxina/farmacología , Piridonas/farmacología , Tubulina (Proteína)/metabolismo , Alquilación , Animales , Sitios de Unión , Bovinos , Colchicina/metabolismo , Yodoacetamida/farmacología , Espectrometría de Fluorescencia , Relación Estructura-Actividad
11.
J Protein Chem ; 17(7): 685-90, 1998 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-9853684

RESUMEN

IKP104 is one of a group of tubulin-binding drugs whose interaction with tubulin suggests that it may bind to the protein at or close to the region where vinblastine binds. By itself IKP104 is a potent enhancer of tubulin decay as evidenced by the fact that it induces the exposure of the sulfhydryl groups and hydrophobic areas on tubulin. In this respect, IKP104 differs from vinblastine and other drugs such as phomopsin A, dolastatin 10, rhizoxin, and maytansine which are competitive or noncompetitive inhibitors of vinblastine binding. In contrast, however, in the presence of colchicine, IKP104 behaves differently and strongly stabilizes tubulin, to an extent much greater than does colchicine alone. IKP104 appears to have two classes of binding site on tubulin, differing in affinity; the acceleration of decay appears to be mediated by the low-affinity site (Chaudhuri et al., 1998, J. Protein Chem., in press). We investigated the relationship of the binding of IKP104 and vinblastine. We found that the high-affinity site or sites of IKP104 overlap with or interact with the vinblastine-binding sites, but that the low-affinity site is distinctly different.


Asunto(s)
Antineoplásicos/metabolismo , Piridonas/metabolismo , Tubulina (Proteína)/metabolismo , Vinblastina/metabolismo , Alquilación , Animales , Sitios de Unión , Química Encefálica , Bovinos , Cinética , Modelos Químicos , Unión Proteica , Espectrometría de Fluorescencia
12.
Biochemistry ; 37(49): 17157-62, 1998 Dec 08.
Artículo en Inglés | MEDLINE | ID: mdl-9860828

RESUMEN

Tubulin, the major subunit protein of microtubules, has a tendency to lose its ability to assemble or to interact with ligands in a time-dependent process known as decay. Decay involves the increase in exposure of sulfhydryl groups and hydrophobic areas. The antimitotic drug IKP104 [2-(4-fluorophenyl)-1-(2-chloro-3, 5-dimethoxyphenyl)-3-methyl-6-phenyl-4(1H)-pyridinone] accelerates the decay of tubulin [Ludueña et al. (1995) Biochemistry 34, 15751-15759]. In the presence of colchicine, however, IKP104 stabilizes tubulin against decay. We have shown that the stability and the acceleration of the decay of tubulin are mediated respectively by the high- and low-affinity binding site(s) of IKP104 [Chaudhuri et al. (1998) J. Protein Chem. 17, 303-309]. To better understand the mechanism by which colchicine protects tubulin from IKP104-induced decay, we examined the effect of colchicine and its analogues on this process. We found that IKP104 unfolds tubulin in a process involving a specific domain where colchicine interacts, although the binding sites of these two drugs are distinctly different. 2-Methoxy-5-(2',3',4'-trimethoxyphenyl) tropolone (MTPT), the bicyclic analogue of colchicine that lacks the B-ring, can also protect tubulin from IKP104-induced decay. An A-ring analogue of colchicine, 3,4,5-trimethoxybenzaldehyde (TMB), can also stop IKP104-induced unfolding of tubulin significantly. Interestingly, the C-ring analogue of colchicine, tropolone methyl ether (TME), does not prevent this process. Our results thus suggest that neither the B-ring nor the C-ring binding regions of colchicine are involved in the IKP104-induced decay and that the A-ring binding site of colchicine on tubulin plays a crucial role in IKP104-induced decay.


Asunto(s)
Colchicina/metabolismo , Piridonas/farmacología , Moduladores de Tubulina , Tubulina (Proteína)/metabolismo , Alquilación/efectos de los fármacos , Animales , Benzaldehídos/farmacología , Sitios de Unión/efectos de los fármacos , Bovinos , Colchicina/análogos & derivados , Colchicina/química , Conformación Proteica/efectos de los fármacos , Piridonas/metabolismo , Espectrometría de Fluorescencia , Tropolona/análogos & derivados , Tropolona/farmacología , Tubulina (Proteína)/química
13.
J Protein Chem ; 17(4): 303-9, 1998 May.
Artículo en Inglés | MEDLINE | ID: mdl-9619583

RESUMEN

IKP104, a novel antimitotic drug, has two classes of binding sites on bovine brain tubulin with different affinities. IKP104, by itself, enhances the decay of tubulin, but in the presence of colchicine or podophyllotoxin, it stabilizes tubulin instead of opening up the hydrophobic areas [Luduena et al. (1995), Biochemistry 34, 15751-15759]. Here, we have dissected these two apparently contradictory effects of IKP104 by cleaving the C-terminal ends of both alpha and beta subunits of tubulin with subtilisin. We have found that the selective removal of the C-terminal ends from both the alpha and beta subunits of alphabeta tubulin lowers the sulfhydryl titer by approximately 1.5 mol/mol of dimer. Interestingly, IKP104 does not increase either the sulfhydryl titer or the exposure of hydrophobic areas of this subtilisin-treated tubulin (alpha(s)beta(s)). Moreover, IKP104 lowers the sulfhydryl titer of alpha(s)beta(s) tubulin approximately by 1 mol/mol and appears to inhibit completely the time-dependent decay of alpha(s)beta(s) tubulin. The cleavage at the C-terminal ends of both alpha and beta modulates the effect of IKP104 on the beta subunit, but not on the alpha subunit. Fluorometric binding data analysis suggests that IKP104 binds to the alpha(s)beta(s) tubulin only at the high-affinity site; the low-affinity site(s) disappear almost completely. The sulfhydryl titer data for alpha and beta and the fluorometric data therefore suggest that the interaction of IKP104 at the high-affinity site on tubulin is not regulated by the C-terminal domains of alpha and beta and the effect of the high-affinity site is restricted largely to the alpha subunit, while the low-affinity-site binding is modulated by the C-terminal domain of beta. It also appears that the stabilization and the acceleration of the decay of tubulin are mediated by distinct interactions of IKP104 with its high- and low-affinity sites on tubulin, respectively.


Asunto(s)
Antineoplásicos/metabolismo , Piridonas/metabolismo , Tubulina (Proteína)/metabolismo , Animales , Sitios de Unión , Encéfalo/metabolismo , Bovinos , Unión Proteica , Espectrometría de Fluorescencia , Tubulina (Proteína)/química
14.
J Protein Chem ; 16(2): 99-105, 1997 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-9112603

RESUMEN

Tubulin, the major component of microtubules, has a tendency to lose its ability to assemble or to bind to ligands in a time-dependent process known as "decay." The decay process also causes tubulin to expose sulfhydryl groups and hydrophobic areas. The antimitotic drug phomopsin A strongly protects the tubulin molecule from decay. Here we have studied the interaction of phomopsin A with alpha beta tubulin and tubulin which has been treated with subtilisin to remove selectively the C-termini of the alpha and beta chains (alpha(s) beta(s)). The binding of phomopsin A to alpha beta tubulin decreases the sulfhydryl titer by approximately 1.0 mol/mol. Selective removal of the peptides from the C-terminal ends does not affect phomopsin A's interaction with tubulin. Moreover, the alpha(s) beta(s) tubulin-phomopsin A complex appears to be more stable than the alpha bet tubulin-phomopsin A complex as determined by the time-dependent increase in exposure of sulfhydryl groups and hydrophobic areas on tubulin. In fact, phomopsin A inhibits the decay process of alpha(s) beta(s) tubulin completely. This observation raises the possibility of determining the conformation of this configuration of tubulin.


Asunto(s)
Química Encefálica , Micotoxinas/metabolismo , Subtilisinas/farmacología , Tubulina (Proteína)/metabolismo , Alquilación , Naftalenosulfonatos de Anilina/metabolismo , Animales , Bovinos , Colorantes Fluorescentes , Estructura Molecular , Micotoxinas/química , Fragmentos de Péptidos/metabolismo , Subtilisinas/metabolismo
17.
Biochem Pharmacol ; 51(7): 903-9, 1996 Apr 12.
Artículo en Inglés | MEDLINE | ID: mdl-8651940

RESUMEN

Griseofulvin is an anti-fungal drug whose mechanism of action is directed against microtubules. Although it inhibits the assembly of mammalian brain tubulin, its binding to tubulin has not been directly measured successfully. We have examined the interaction of griseofulvin with tubulin fluorometrically by measuring the quenching of tubulin tryptophan fluorescence by griseofulvin. From Scatchard analysis, we found that griseofulvin bound to tubulin at one class of binding site with an affinity constant of 1.2 +/- 0.19 x 10(4) M(-1), and the binding was largely reversible. Griseofulvin caused a major change in the conformation of tubulin in that it increased the sulfhydryl titer of tubulin approximately 2-fold. The drug affected both the alpha and beta subunits of tubulin equally. Interestingly, griseofulvin did not increase the sulfhydryl titer of the tubulin-colchicine complex although the binding site of griseofulvin was distinctly different from that of colchicine. The change of conformation of tubulin upon interaction with griseofulvin did not affect the exposure of hydrophobic areas on tubulin as shown by binding of bis-5,5'-[8(N-phenyl)aminonapthalene-1-sulfonic acid] (BisANS). Even in combination with colchicine, griseofulvin had very little effect on BisANS binding to tubulin. Thus, griseofulvin appears to interact with tubulin in a manner that is very different from that of many other tubulin ligands.


Asunto(s)
Encéfalo/efectos de los fármacos , Griseofulvina/farmacología , Tubulina (Proteína)/efectos de los fármacos , Alquilación/efectos de los fármacos , Animales , Bovinos , Colchicina/farmacología , Relación Dosis-Respuesta a Droga , Cinética
18.
Biochemistry ; 34(48): 15751-9, 1995 Dec 05.
Artículo en Inglés | MEDLINE | ID: mdl-7495806

RESUMEN

The ligands of tubulin have proved to be excellent probes for the conformation of the tubulin molecule. The most varied in their effects on tubulin are those ligands which are competitive or noncompetitive inhibitors of vinblastine binding. The 4(H)-pyrizinone derivative 2-(4-fluorophenyl)-1-(2-chloro- 3,5-dimethoxyphenyl)-3-methyl-6-phenyl-4(1H)-pyridinone [sequence: see text] (IKP104) is a novel antimitotic drug which inhibits microtubule assembly in vitro and in vivo and polymerizes tubulin into spiral filaments. Using a fluorescence assay, we found that IKP104 appears to bind to tubulin at two classes of site, differing in affinity. IKP104 also blocks formation of an intrachain cross-link in beta-tubulin, induced by N,N"-ethylenebis(iodoacetamide), linking Cys12 to either Cys201 or Cys211. IKP104 appears to belong to that group of tubulin ligands which includes vinblastine, maytansine, rhizoxin, phomopsin A, dolastatin 10, and halichondrin B. An unusual effect of IKP104 is that it greatly enhances the decay or apparent unfolding or opening of the tubulin molecule. The sulfhydryl titer of tubulin is doubled and the exposure of hydrophobic areas on the tubulin molecule is tripled by IKP104. These effects of IKP104 are counteracted by vinblastine, maytansine, and phomopsin A, suggesting that IKP104 may be competing with these other drugs for binding to tubulin. However, the effects are also counteracted by colchicine and podophyllotoxin, implying a more complex effect, namely, that IKP104 and colchicine, even when both are bound to tubulin, are competing for their effects on the same domain of tubulin. Surprisingly, when IKP104 is used in conjunction with colchicine, binding of colchicine to tubulin is strongly stabilized.(ABSTRACT TRUNCATED AT 250 WORDS)


Asunto(s)
Antineoplásicos/metabolismo , Encéfalo/metabolismo , Piridonas/metabolismo , Tubulina (Proteína)/metabolismo , Alquilación , Animales , Antineoplásicos/farmacología , Radioisótopos de Carbono , Bovinos , Colchicina/metabolismo , Yodoacetamida/química , Mitosis/efectos de los fármacos , Unión Proteica , Conformación Proteica/efectos de los fármacos , Piridonas/farmacología , Compuestos de Sulfhidrilo/química , Tritio , Tubulina (Proteína)/química
19.
Prep Biochem ; 24(3-4): 185-92, 1994 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-7831201

RESUMEN

The phenomenon of differential charge distribution on sperm surface membrane has been utilised here in a low e.m.f. (electro motive force) capillary electrophoresis system to effect separation of sperm heads from disintegrated mixed spermatozoal subfractions. Washed caudal sperm of goat (Black Bengal variety) and ejaculated washed human sperm were fractionated by sonication into head, mid-piece and tail portions. Routine techniques of density gradient centrifugation on Percoll and/or sucrose were performed with sonicated spermatozoa for separation into their respective subfractions. The products obtained were not free of contamination in either case. Mixed sperm fractions when subjected to the afore mentioned modified capillary electrophoresis technique only the head pieces exhibited high affinity for migration towards the cathode terminal. With this method around 50% of the total sperm heads were separated and collected in absolutely pure form at the cathode side within 2 hrs. at 150 volts (V) and 1.5 milliampere (mA) current at 37.5 degrees C. A 4 cm. long capillary tube with a bore size 1.2 mm. was used for this purpose.


Asunto(s)
Separación Celular/métodos , Electroforesis , Cabeza del Espermatozoide , Animales , Centrifugación por Gradiente de Densidad , Cabras , Humanos , Masculino , Povidona , Dióxido de Silicio , Sonicación
20.
Biochem Mol Biol Int ; 29(3): 421-8, 1993 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-8485459

RESUMEN

The unique property of Mimosa pudica microtubules is that they are cold stable in nature. They do not dissociate at 0 degrees C unlike animal microtubules. No other protein fractions of M. pudica are responsible for the cold-resistance property of these microtubules. It seems that tubulin, itself, has an intrinsic cold resistance which makes it unique compared with animal tubulin. Moreover, M. pudica microtubule makes cold-sensitive goat brain microtubule cold-resistant when used in greater than the suboptimal concentration.


Asunto(s)
Microtúbulos/fisiología , Proteínas de Plantas/metabolismo , Tubulina (Proteína)/fisiología , Animales , Encéfalo/metabolismo , Frío , Cabras , Tubulina (Proteína)/farmacología
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...