Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 38
Filtrar
Mais filtros

Base de dados
Tipo de documento
País de afiliação
Intervalo de ano de publicação
1.
Biochim Biophys Acta ; 1832(10): 1634-52, 2013 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-23628734

RESUMO

The "amyloid-ß (Aß) hypothesis" posits that accumulating Aß peptides (Aßs) produced by neurons cause Alzheimer's disease (AD). However, the Aßs contribution by the more numerous astrocytes remains undetermined. Previously we showed that fibrillar (f)Aß25-35, an Aß42 proxy, evokes a surplus endogenous Aß42 production/accumulation in cortical adult human astrocytes. Here, by using immunocytochemistry, immunoblotting, enzymatic assays, and highly sensitive sandwich ELISA kits, we investigated the effects of fAß25-35 and soluble (s)Aß25-35 on Aß42 and Aß40 accumulation/secretion by human cortical astrocytes and HCN-1A neurons and, since the calcium-sensing receptor (CaSR) binds Aßs, their modulation by NPS 2143, a CaSR allosteric antagonist (calcilytic). The fAß25-35-exposed astrocytes and surviving neurons produced, accumulated, and secreted increased amounts of Aß42, while Aß40 also accrued but its secretion was unchanged. Accordingly, secreted Aß42/Aß40 ratio values rose for astrocytes and neurons. While slightly enhancing Aß40 secretion by fAß25-35-treated astrocytes, NPS 2143 specifically suppressed the fAß25-35-elicited surges of endogenous Aß42 secretion by astrocytes and neurons. Therefore, NPS 2143 addition always kept Aß42/Aß40 values to baseline or lower levels. Mechanistically, NPS 2143 decreased total CaSR protein complement, transiently raised proteasomal chymotrypsin activity, and blocked excess NO production without affecting the ongoing increases in BACE1/ß-secretase and γ-secretase activity in fAß25-35-treated astrocytes. Compared to fAß25-35, sAß25-35 also stimulated Aß42 secretion by astrocytes and neurons and NPS 2143 specifically and wholly suppressed this effect. Therefore, since NPS 2143 thwarts any Aß/CaSR-induced surplus secretion of endogenous Aß42 and hence further vicious cycles of Aß self-induction/secretion/spreading, calcilytics might effectively prevent/stop the progression to full-blown AD.


Assuntos
Doença de Alzheimer/metabolismo , Peptídeos beta-Amiloides/metabolismo , Astrócitos/efeitos dos fármacos , Naftalenos/farmacologia , Neurônios/efeitos dos fármacos , Receptores de Detecção de Cálcio/antagonistas & inibidores , Adulto , Doença de Alzheimer/patologia , Doença de Alzheimer/terapia , Peptídeos beta-Amiloides/fisiologia , Astrócitos/metabolismo , Biopterinas/análogos & derivados , Biopterinas/farmacologia , Células Cultivadas , Humanos , Naftalenos/uso terapêutico , Neurônios/metabolismo
2.
Cells ; 12(12)2023 06 13.
Artigo em Inglês | MEDLINE | ID: mdl-37371088

RESUMO

The enormous, 2-3-million-year evolutionary expansion of hominin neocortices to the current enormity enabled humans to take over the planet. However, there appears to have been a glitch, and it occurred without a compensatory expansion of the entorhinal cortical (EC) gateway to the hippocampal memory-encoding system needed to manage the processing of the increasing volume of neocortical data converging on it. The resulting age-dependent connectopathic glitch was unnoticed by the early short-lived populations. It has now surfaced as Alzheimer's disease (AD) in today's long-lived populations. With advancing age, processing of the converging neocortical data by the neurons of the relatively small lateral entorhinal cortex (LEC) inflicts persistent strain and high energy costs on these cells. This may result in their hyper-release of harmless Aß1-42 monomers into the interstitial fluid, where they seed the formation of toxic amyloid-ß oligomers (AßOs) that initiate AD. At the core of connectopathic AD are the postsynaptic cellular prion protein (PrPC). Electrostatic binding of the negatively charged AßOs to the positively charged N-terminus of PrPC induces hyperphosphorylation of tau that destroys synapses. The spread of these accumulating AßOs from ground zero is supported by Aß's own production mediated by target cells' Ca2+-sensing receptors (CaSRs). These data suggest that an early administration of a strongly positively charged, AßOs-interacting peptide or protein, plus an inhibitor of CaSR, might be an effective AD-arresting therapeutic combination.


Assuntos
Doença de Alzheimer , Humanos , Doença de Alzheimer/metabolismo , Peptídeos beta-Amiloides/metabolismo , Hipocampo/metabolismo , Neurônios/metabolismo , Córtex Entorrinal/metabolismo , Proteínas Priônicas/metabolismo
3.
Biochim Biophys Acta ; 1783(9): 1642-53, 2008 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-18439918

RESUMO

Lamin B1, a major component of the nuclear lamina, anchors the nucleus to the cytoskeletal cage, and controls nuclear orientation, chromosome positioning and, alongside several enzymes, fundamental nuclear functions. Exposing polyomavirus-transformed rat pyF111 fibroblasts and human cervical carcinoma (HCC) C4-I cells for 30 min to photoexcited perylenequinone calphostin C, i.e. Cal C(phiE), an established reactive oxygen species (ROS)-generator and protein kinase C (PKC) inhibitor, caused the cells to selectively oxidize and then totally destroy their nuclear lamin B1 by only 60 min after starting the treatment, i.e. when apoptotic caspases' activities had not yet increased. However, while the oxidized lamin B1 was being destroyed, lamins A/C, the lamin A-associated nuclear envelope protein emerin, and the nucleoplasmic protein cyclin E were neither oxidized nor destroyed. The oxidized lamin B was ubiquitinated and demolished in the proteasome probably by an enhanced peptidyl-glutaminase-like activity. Hence, the Cal C(phiE)-induced rapid and selective lamin B1 oxidation and proteasomal destruction ahead of the activation of apoptotic caspases was by itself a most severe molecular lesion impairing vital nuclear functions. Conversely, Cal C directly added to the cells kept in the dark damaged neither nuclear lamin B1 nor cell viability. Thus, our findings reveal a novel cell-damaging mechanism of a photodynamic tumor therapeutic agent.


Assuntos
Antibióticos Antineoplásicos/farmacologia , Lamina Tipo B/metabolismo , Naftalenos/farmacologia , Neoplasias/metabolismo , Animais , Antibióticos Antineoplásicos/efeitos da radiação , Apoptose , Linhagem Celular Transformada , Linhagem Celular Tumoral , Humanos , Imuno-Histoquímica , Lamina Tipo B/análise , Lamina Tipo B/imunologia , Naftalenos/efeitos da radiação , Neoplasias/patologia , Membrana Nuclear/metabolismo , Estresse Oxidativo , Fotoquimioterapia , Complexo de Endopeptidases do Proteassoma/metabolismo , Ratos , Ubiquitinação
4.
Curr Pharm Des ; 23(33): 4990-5000, 2017.
Artigo em Inglês | MEDLINE | ID: mdl-28699522

RESUMO

Understanding the pathophysiology of Alzheimer's disease (AD) in the principal human neural cells is necessary for finding therapeutics for this illness. To help do this, we have been using freshly cultured functionally normal cerebral cortical adult human astrocytes (NAHAs) and postnatal neurons. The findings show that amyloid-ß oligomers (Aß-os) binding to calcium-sensing receptors (CaSRs) on NAHAs and neuron surfaces trigger signals capable of driving AD pathogenesis. This Aß•CaSR signalling shifts the amyloid precursor protein (APP) from its α-secretase shedding producing neurotrophic/neuroprotective soluble (s)APPα to its ß-secretase cleaving engendering AD-driving Aß42/Aß42-os peptides. Aß•CaSR signalling in NAHAs also drives the release of toxic hyper-phosphorylated Tau proteins in exosomes, and of nitric oxide, and VEGF-A. These several harmful agents comprise the neuron-killing machinery, driving the very slowly spreading AD neurocontagion. VEGF-A over-secretion from Aß-exposed blood vessel-attached astrocytes induces a functional magnetic resonance imaging- detectable hippocampal neoangiogenesis which indicates approaching AD in amnestic minor cognitive impairment (aMCI) patients. Most important in AD's regard, selective allosteric CaSR antagonists (calcylitics) added to Aß42/Aß42-os-exposed NAHAs (or to human neuron cultures) rescue the extracellular shedding of neurotrophic/ neuroprotective sAPPα and suppress all the neurotoxic effects of Aß•CaSR signalling even when multiple microglial cytokines are also present. Therefore, since the multipotent calcilytics would be reasonably safe and inexpensive drugs for humans, it is worthwhile testing them as AD therapeutics in clinical trials especially in persons in the earliest detectable stages of AD neuropathology progression such as aMCI.


Assuntos
Doença de Alzheimer/tratamento farmacológico , Doença de Alzheimer/metabolismo , Astrócitos/metabolismo , Sistemas de Liberação de Medicamentos/tendências , Receptores de Detecção de Cálcio/antagonistas & inibidores , Receptores de Detecção de Cálcio/metabolismo , Animais , Astrócitos/efeitos dos fármacos , Bloqueadores dos Canais de Cálcio/administração & dosagem , Sistemas de Liberação de Medicamentos/métodos , Humanos , Resultado do Tratamento
5.
Cancer Lett ; 244(1): 8-15, 2006 Nov 28.
Artigo em Inglês | MEDLINE | ID: mdl-16540235

RESUMO

Between 1958 and the late 1970s it was learned that PTH (the parathyroid hormone) could directly stimulate the initiation of DNA replication by murine CFU-S (colony-forming unit-spleen) cells via cyclic AMP, stimulate the proliferation of normal and X-irradiated murine and rat bone marrow cells, control hematopoiesis, and increase the survival of X-irradiated mice and rats when injected any time between 18h before and 3h after X-irradiation. Since then, it has been shown that the hematopoietic stem cell niche consists of PTH receptor-bearing, osteoblastic trabecular bone-lining cells that maintain the stem cells' (HSCs') proliferatively quiescent 'stemness' by various gene up-regulating and down-regulating signals caused by the tight adhesion of the HSCs to the osteoblastic niche-lining cells. Stimulating the osteoblastic lining cells with recombinant human PTH-(1-34) (Forteo) causes a cyclic AMP-mediated enlargement of the HSC pool and promotes bone marrow transplant engraftment and growth and the survival of lethally irradiated mice. But this is only the beginning of the exploitation of the PTHs for marrow engraftment. It must now be determined whether the marrow engraftment-enhancing action of this potent bone growth-stimulating PTH can be extended from mice to rats and monkeys. It must be determined whether two other PTH peptides, rhPTH-(1-84) [Preos]and [Leu(27)]cyclo(Glu(22)-Lys(26))hPTH-(1-31)NH(2) [Ostabolin-C]) are as effective as or better than rhPTH-(1-34)(Forteo). Since, all three peptides are on the market, or nearing the market, for safely and strongly stimulating bone growth and treating osteoporosis one or all of them may become valuable tools for safely promoting the engraftment of peripherally harvested HSCs in cancer patients whose bone marrows have been 'emptied' by chemotherapeutic drugs or ionizing radiation.


Assuntos
Antineoplásicos/efeitos adversos , Medula Óssea/efeitos dos fármacos , Hematopoese/efeitos dos fármacos , Hormônio Paratireóideo/farmacologia , Radiação Ionizante , Animais , Medula Óssea/efeitos da radiação , Hematopoese/efeitos da radiação , Humanos , Neoplasias/tratamento farmacológico , Neoplasias/radioterapia
6.
Curr Opin Investig Drugs ; 7(4): 349-59, 2006 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-16625822

RESUMO

The first experiments demonstrating parathyroid hormone's (PTH's) dramatic bone-building activity in rat pups, using a bovine parathyroid extract called parathormone were reported 74 years ago. Over the next decades, the native parathyroid hormone (human (h)PTH(1-84)) was purified and two of its fragments (hPTH(1-34) and (Leu27)cycloGlu22-Lys26hPTH(1-31)NH2) have been developed for the treatment of osteoporosis. One of these, recombinant (r)hPTH(1-34), is now on the market under the trade name of Forteo. The native hormone has also completed clinical trials and (Leu27)cycloGlu22-Lys26hPTH(1-31)NH2 is in phase II clinical trials under the trade name Ostabolin-C. All three of these peptides potently stimulate bone growth, reinforce bone microstructure weakened by estrogen deprivation and reduce further fracturing. Furthermore, future studies may demonstrate that these peptides also promote the repair of existing fractures and implant anchorage in both healthy and osteoporotic humans. PTHs have the potential to become more successful by using cost-cutting, but still effective, cyclical treatment regimens and by formulating them for non-injectable delivery. This review will discuss the identification of PTH peptides, how they function and their future role in the treatment of osteoporosis.


Assuntos
Osteoporose/tratamento farmacológico , Hormônio Paratireóideo/uso terapêutico , Animais , Densidade Óssea/efeitos dos fármacos , Desenvolvimento Ósseo/efeitos dos fármacos , Quinase 3 da Glicogênio Sintase/fisiologia , Glicogênio Sintase Quinase 3 beta , Humanos , Osteoporose/fisiopatologia , Hormônio Paratireóideo/farmacologia , Fragmentos de Peptídeos/uso terapêutico , Receptor Tipo 1 de Hormônio Paratireóideo/fisiologia , Tomografia Computadorizada por Raios X , beta Catenina/fisiologia
7.
Int J Mol Med ; 17(1): 111-20, 2006 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-16328019

RESUMO

Protein kinase C (PKC) isoforms regulate cell proliferation and apoptosis. Since the PKC isoenzyme complement varies considerably from cell type to cell type, a PKC's responsiveness to an apoptogenic challenge must be defined for both the type of apoptogen and the type of cell. We have already reported that the changes in the distribution and activity of PKC-delta in apoptosing polyomavirus-infected/transformed Fischer rat embryo pyF111 fibroblasts depend on the type of apoptogen. Here, we show that this is also true for PKC-betaI in pyF111 cells treated with the slow DNA-damaging VP-16 (etoposide) or the fast-acting (in the cytoplasm) calphostin C. These apoptogens caused quite different shifts of the PKC-betaI level and activity in the nuclear membrane (NM) and nucleoplasm (NP), but corresponding changes in the cytosol (CS) and cytoplasmic particulate (CP) fractions. The hefty translocation of PKC-betaI onto the CP fraction and its increased activity there suggest the possible triggering of a cytochrome c/caspase-mediated apoptosis-inducing mechanism common to both agents. The present results are a necessary lead-up to functional proteomic analyses aimed at identifying the molecules forming the local PKC-betaI signalling modules under different conditions.


Assuntos
Transformação Celular Viral , Etoposídeo/metabolismo , Fibroblastos/metabolismo , Naftalenos/metabolismo , Polyomavirus/metabolismo , Proteína Quinase C/metabolismo , Animais , Apoptose/fisiologia , Linhagem Celular , Núcleo Celular/enzimologia , Citoplasma/enzimologia , Inibidores Enzimáticos/metabolismo , Fibroblastos/citologia , Isoenzimas/metabolismo , Inibidores da Síntese de Ácido Nucleico/metabolismo , Polyomavirus/genética , Proteína Quinase C beta , Ratos , Ratos Endogâmicos F344
8.
Int J Mol Med ; 16(5): 801-7, 2005 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-16211247

RESUMO

The accumulation of amyloid beta (Abeta)-peptides and their collection in fibrillar plaques in the human brain are believed to be responsible for Alzheimer's disease. The major neuron killers in the Alzheimer brain include proinflammatory cytokines and NO made by NOS-2 (inducible nitric oxide synthase-2). We have determined the effect of a soluble Abeta peptide, Abeta(1-40), on the expression of NOS-2 in astrocytes using a novel model system consisting of pure cultures of cells from adult human brains that, after the first three passages in vitro, become stably locked into the normal astrocytic phenotype like their counterparts in the adult human brain. Abeta(1-40) alone stimulated quiescent astrocytes to start expressing functional NOS-2 and dumping NO into the culture medium during the next 4 days. But adding three of the proinflammatory cytokines commonly produced in the Alzheimer brain--IFN-gamma, IL-1beta, and TNF-alpha--along with Abeta(1-40) more than trebled NOS-2 expression and doubled NO production. In view of the possibility of myelin breakdown in the Alzheimer brain, we also tested the capability of myelin basic protein (MBP) to stimulate NO production using human astrocytes. We found that MBP mimicked the ability of Abeta(1-40) to induce cells to release NO and adding the cytokine triad along with MBP more than doubled NO production and release. Thus, it appears that Abeta peptides and MBP can join forces with proinflammatory cytokines to enhance the NO-mediated killing of neurons in the Alzheimer brain.


Assuntos
Doença de Alzheimer/etiologia , Peptídeos beta-Amiloides/farmacologia , Astrócitos/enzimologia , Citocinas/farmacologia , Proteína Básica da Mielina/farmacologia , Fragmentos de Peptídeos/farmacologia , Adulto , Doença de Alzheimer/enzimologia , Astrócitos/efeitos dos fármacos , Células Cultivadas , Sinergismo Farmacológico , Humanos , Óxido Nítrico/metabolismo
9.
Cells ; 4(3): 253-74, 2015 Jul 13.
Artigo em Inglês | MEDLINE | ID: mdl-26184316

RESUMO

Dentate-gyral granule cells in the hippocampus plus dentate gyrus memory-recording/retrieving machine, unlike most other neurons in the brain, are continuously being generated in the adult brain with the important task of separating overlapping patterns of data streaming in from the outside world via the entorhinal cortex. This "adult neurogenesis" is driven by tools in the mature granule cell's cilium. Here we report our discovery of leptin's LepRb receptor in this cilium. In addition, we discuss how ciliary LepRb signaling might be involved with ciliary p75NTR and SSTR3 receptors in adult neurogenesis and memory formation as well as attenuation of Alzheimer's neuropathology by reducing the production of its toxic amyloid-ß-derived drivers.

10.
Curr Pharm Biotechnol ; 16(8): 696-706, 2015.
Artigo em Inglês | MEDLINE | ID: mdl-25941885

RESUMO

The "amyloid cascade hypothesis" posits that an extracellular build-up of amyloid-ß oligomers (Aß-os) and polymers (fibrils) subsequently inducing toxic hyperphosphorylated (p)-Tau oligomers (p-Tau-os) and neurofibrillary tangles starts the sporadic late-onset Alzheimer's disease (LOAD) in the aged lateral entorhinal cortex. Conversely, mutated genes cause a diffuse cerebral Aßs/Aß-os overproduction promoting early-onset familiar AD (EOFAD). Surplus exogenous Aß-os exert toxic actions at several levels. They reach the nuclei of human astrocyte-neurons teams (ANTs) to enhance the transcription of Aß precursor protein (APP) and ß-secretase/BACE1 genes. The overexpressed APP and BACE1 proteins act in concert with γ-secretase to overproduce endogenous Aßs/Aß-os, of which a few enter the nuclei to upkeep Aßs overproduction, while the rest gather in the cytoplasm, damage mitochondria, and are oversecreted. Simultaneously, extracellular Aß-os bind the ANTs' calcium-sensing receptors (CaSRs) activating signalings that hinder the proteolysis and hence favor the surplus hoarding/secretion of Aßs/Aß-os. Overreleased Aß-os spread, reach growing numbers of adjacent ANTs to recruit them to overproduce/oversecrete further Aß-os amounts via the just mentioned mechanisms. Alongside, Aß•CaSR signalings elicit a noxious overproduction/overrelease of nitric oxide (NO) and vascular endothelial growth factor (VEGF)-A from ANTs' astrocytes. While astrocytes survive the toxic onslaught, neurons die. Thus, AD progression is driven by ceaselessly self-sustaining neurotoxic cycles, which engender first Aß-os and later p-Tau-os that cooperatively destroy increasingly wider cognition-related cortical areas. Notably, a highly selective allosteric CaSR antagonist (calcilytic), like NPS 2143, does preserve human cortical postnatal HCN-1A neurons viability notwithstanding the presence of exogenous Aß-os by suppressing the otherwise elicited oversecretion and spread of newly synthesized Aß-os. Therefore, if given at minimal cognitive impairment or earlier stages, calcilytics could halt AD progression and preserve the patients' cortical neurons, cognitive abilities, and eventually life.


Assuntos
Doença de Alzheimer/prevenção & controle , Cálcio/metabolismo , Doença de Alzheimer/metabolismo , Doença de Alzheimer/patologia , Secretases da Proteína Precursora do Amiloide/metabolismo , Peptídeos beta-Amiloides/metabolismo , Animais , Astrócitos/metabolismo , Humanos , Proteínas tau/metabolismo
11.
Neuroscientist ; 21(1): 9-29, 2015 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-24740577

RESUMO

Evidence has begun emerging for the "contagious" and destructive Aß42 (amyloid-beta42) oligomers and phosphorylated Tau oligomers as drivers of sporadic Alzheimer's disease (AD), which advances along a pathway starting from the brainstem or entorhinal cortex and leading to cognition-related upper cerebral cortex regions. Seemingly, Aß42 oligomers trigger the events generating the neurotoxic Tau oligomers, which may even by themselves spread the characteristic AD neuropathology. It has been assumed that only neurons make and spread these toxic drivers, whereas their associated astrocytes are just janitorial bystanders/scavengers. But this view is likely to radically change since normal human astrocytes freshly isolated from adult cerebral cortex can be induced by exogenous Aß25-35, an Aß42 proxy, to make and secrete increased amounts of endogenous Aß42. Thus, it would seem that the steady slow progression of AD neuropathology along specific cognition-relevant brain networks is driven by both Aß42 and phosphorylated Tau oligomers that are variously released from increasing numbers of "contagion-stricken" members of tightly coupled neuron-astrocyte teams. Hence, we surmise that stopping the oversecretion and spread of the two kinds of "contagious" oligomers by such team members, perhaps via a specific CaSR (Ca(2+)-sensing receptor) antagonist like NPS 2143, might effectively treat AD.


Assuntos
Doença de Alzheimer/etiologia , Doença de Alzheimer/metabolismo , Peptídeos beta-Amiloides/metabolismo , Astrócitos/metabolismo , Encéfalo/metabolismo , Neurônios/metabolismo , Fragmentos de Peptídeos/metabolismo , Proteínas tau/metabolismo , Doença de Alzheimer/patologia , Animais , Astrócitos/patologia , Encéfalo/patologia , Humanos , Neurônios/patologia , Fosforilação
12.
Crit Rev Eukaryot Gene Expr ; 12(1): 23-51, 2002.
Artigo em Inglês | MEDLINE | ID: mdl-12433064

RESUMO

There is a need for anabolic drugs that can stimulate bone growth, improve bone microarchitecture, accelerate fracture healing and thus restore bone strength to oteoporotics. The anabolic agents currently leading the way to the clinic are the parathroid hormone (PTH) and some of its adenylyl cyclase-stimulating fragments. Here we discuss what is known about the genes and their products that are stimulated by PTHR1 receptor signals and in four ways cause a large accumulation of bone-building osteoblasts. We will also discuss the currently controversial anabolic activity of the cholesterol-lowering statins and outline a possible mechanism by which they might stimulate BMP-2 expession and bone growth. Finally, we will present the growing evidence for the body's "fat-o-stat" cytokine-leptin-indirectly restraining bone growth via a hypothalamic factor and at the same serving as a local autocrine/paracrine stimulator of osteoblast activity via IGF-I and an inhibitor of osteoclast generation by stimulating osteoblastic cells' antiosteoclast OPG (osteoprotegerin) expression and reducing their proosteoclast RANKL expression.


Assuntos
Desenvolvimento Ósseo/efeitos dos fármacos , Desenvolvimento Ósseo/fisiologia , Inibidores de Hidroximetilglutaril-CoA Redutases/farmacologia , Leptina/fisiologia , Hormônio Paratireóideo/farmacologia , Animais , Cisteína Endopeptidases/efeitos dos fármacos , Inibidores de Cisteína Proteinase/farmacologia , Humanos , Complexos Multienzimáticos/efeitos dos fármacos , Complexo de Endopeptidases do Proteassoma
13.
Curr Med Chem ; 11(21): 2867-81, 2004 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-15544480

RESUMO

Osteogenic peptides are, or have potential to be, therapies for the treatment of osteoporosis, fracture repair, and repair of loosened bone implants. Human parathyroid hormone has been approved for the treatment of post-menopausal osteoporosis. Constrained analogs of PTH and the parathyroid-hormone related peptide (PTHrP) have aided the understanding of how PTH and PTHrP bind to their common receptor and some of these analogs have improved properties that make them possible candidates for clinical trial. Cyclization by lactam formation has shown that a core region of human PTH (hPTH) from residues 16-26 binds as an alpha-helix to the receptor and that the biological effects are remarkably sensitive to ring size. Appropriate cyclization in this region of the molecule not only has yielded analogs with improved receptor activation but also ones less susceptible to protease degradation and thus more active in vivo. Cyclization has been less successful in the N-terminus region, residues 1-12, of hPTH(1-34) with only a cyclization between residues 6 and 10 showing some promise. The growing understanding of how this region binds to the receptor will lead to other productive constraints. This review also covers the potential of a different class of molecule, the osteogenic growth peptide (OPG), as an anabolic bone agent. These molecules have much weaker anabolic effects than PTH and cyclization does not result in improved activity. However, the information gained from these studies may yield analogs with better pharmacological profiles.


Assuntos
Osteogênese/efeitos dos fármacos , Hormônio Paratireóideo/análogos & derivados , Hormônio Paratireóideo/farmacologia , Sequência de Aminoácidos , Animais , Histonas , Humanos , Peptídeos e Proteínas de Sinalização Intercelular/química , Peptídeos e Proteínas de Sinalização Intercelular/farmacologia , Dados de Sequência Molecular , Hormônio Paratireóideo/química , Hormônio Paratireóideo/uso terapêutico , Receptores de Hormônios Paratireóideos/metabolismo
14.
Treat Endocrinol ; 1(3): 175-90, 2002.
Artigo em Inglês | MEDLINE | ID: mdl-15799210

RESUMO

The susceptibility to traumatic fracturing of osteopenic bones, and the spontaneous fracturing of osteoporotic bones by normal body movements caused by the microstructural deterioration and loss of bone, are currently treated with antiresorptive drugs, such as the bisphosphonates, calcitonin, estrogens, and selective estrogen receptor modulators. These antiresorptive agents target osteoclasts and, as their name indicates, reduce or stop bone resorption. They cannot directly stimulate bone formation, increase bone mass above normal values in ovariectomized rat models, or improve microstructure. However, there is a family of agents - the parathyroid hormone (PTH) and some of its fragments and their analogs - which directly stimulate bone growth and improve microstructure independently from impairing osteoclasts. These drugs are about to make their clinical debut in treating patients with osteoporosis and, probably not too far in the future, for accelerating fracture healing. They stimulate osteoblast accumulation and bone formation in three ways via signals from the type 1 PTH/PTH-related protein (PTHR1) receptors on proliferatively inactive preosteoblasts, osteoblasts, osteocytes and bone-lining cells. The receptor signals shut down the proliferative machinery in preosteoblasts and push their maturation to osteoblasts, cause the osteoblastic cells to make and secrete several factors that stimulate the extensive proliferation of osteoprogenitors without PTHRI receptors, stimulate the reversion of bone-lining cells to osteoblasts, and extend osteoblast lifespan and productivity by preventing them from suicidally initiating apoptosis. The first of the PTHs to reach the clinic will be teriparatide [recombinant human (h)PTH-(1-34)], which was recommended for approval in 2001 by the US Food and Drug Administration Endocrinology and Metabolic Drugs Advisory Committee for the treatment of postmenopausal osteoporosis. Teriparatide has been shown to considerably increase cancellous and cortical bone mass, improve bone microstructure, prevent fractures and thus provide benefits that cannot be provided by current antiresorptive drugs, when administered subcutaneously at a daily dose of 20 microg for no longer than 2 years to patients with osteoporosis.


Assuntos
Osteoporose/tratamento farmacológico , Hormônio Paratireóideo/análogos & derivados , Hormônio Paratireóideo/uso terapêutico , Animais , Densidade Óssea/efeitos dos fármacos , Remodelação Óssea/efeitos dos fármacos , Ensaios Clínicos como Assunto , Terapia de Reposição de Estrogênios , Feminino , Humanos , Masculino , Osteoblastos/metabolismo , Osteoclastos/metabolismo , Osteoporose/patologia , Fragmentos de Peptídeos/uso terapêutico , Ratos , Receptores de Hormônios Paratireóideos/metabolismo , Teriparatida/uso terapêutico
15.
Neuromolecular Med ; 16(4): 645-57, 2014 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-24948534

RESUMO

The excess vascular endothelial growth factor (VEGF) produced in the Alzheimer's disease (AD) brain can harm neurons, blood vessels, and other components of the neurovascular units (NVUs). But could astrocytes partaking in networks of astrocyte-neuron teams and connected to blood vessels of NVUs contribute to VEGF production? We have shown with cultured cerebral cortical normal (i.e., untransformed) adult human astrocytes (NAHAs) that exogenous amyloid-ß peptides (Aßs) stimulate the astrocytes to make and secrete large amounts of Aßs and nitric oxide by a mechanism mediated through the calcium-sensing receptor (CaSR). Here, we report that exogenous Aßs stimulate the NAHAs to produce and secrete even VEGF-A through a CaSR-mediated mechanism. This is indicated by the ability of Aßs to specifically bind the CaSR, and the capability of a CaSR activator, the "calcimimetic" NPS R-568, to imitate, and of the CaSR antagonist, "calcilytic" NPS 2143, to inhibit, the Aßs stimulation of VEGF-A production and secretion by the NAHAs. Thus, Aßs that accumulate in the AD brain may make the astrocytes that envelop and functionally collaborate with neurons into multi-agent AD-driving "machines" via a CaSR signaling mechanism(s). These observations suggest the possibility that CaSR allosteric antagonists such as NPS 2143 might impede AD progression.


Assuntos
Peptídeos beta-Amiloides/farmacologia , Astrócitos/efeitos dos fármacos , Fragmentos de Peptídeos/farmacologia , Receptores de Detecção de Cálcio/fisiologia , Fator A de Crescimento do Endotélio Vascular/biossíntese , Adulto , Regulação Alostérica , Doença de Alzheimer/metabolismo , Peptídeos beta-Amiloides/metabolismo , Compostos de Anilina/farmacologia , Astrócitos/metabolismo , Cálcio/agonistas , Comunicação Celular , Células Cultivadas , Humanos , Subunidade alfa do Fator 1 Induzível por Hipóxia/fisiologia , Naftalenos/farmacologia , Neurônios/metabolismo , Óxido Nítrico/metabolismo , Fenetilaminas , Propilaminas , Ligação Proteica , Receptores de Detecção de Cálcio/antagonistas & inibidores , Lobo Temporal/citologia , Fator A de Crescimento do Endotélio Vascular/genética , Fator A de Crescimento do Endotélio Vascular/metabolismo
16.
Int J Mol Med ; 31(1): 3-10, 2013 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-23124509

RESUMO

The pathophysiological mechanisms underlying the onset and inexorable progression of the late­onset form of Alzheimer's disease (AD) are still the object of controversy. This review takes stock of some most recent advancements of this field concerning the complex roles played by the amyloid­ß (Aß)­binding p75 neurotrophin receptor (p75NTR) and calcium­sensing receptor (CaSR) and by the primary cilia in AD. Apart from their physiological roles, p75NTR is more intensely expressed in the hippocampus of human AD brains and Aß­bound p75NTR triggers cell death, whereas Aß­bound CaSR signalling induces the de novo synthesis and release of nitric oxide (NO), vascular endothelial growth factor (VEGF)­A and Aß peptides (Aßs), particularly on the part of normal adult human astrocytes. The latter effect could significantly increase the pool of Aß­ and NO­producing nerve cells favouring the progressive spread of a self­sustaining and self­reinforcing 'infectious' mechanism of neural and vascular (i.e. blood-brain barrier) cell damage. Interestingly, primary cilia concentrate p75NTR receptors in their membranes and are abnormally structured/damaged in transgenic (Tg) AD­model mice, which could impact on the adult neurogenesis occurring in the dentate gyrus's subgranular zone (SGZ) that is necessary for new memory encoding, thereby favouring typical AD cognitive decline. Altogether, these findings may pave the way to novel therapeutic approaches to AD, particularly in its mild cognitive impairment (MCI) and pre­MCI stages of development.


Assuntos
Doença de Alzheimer/fisiopatologia , Astrócitos/metabolismo , Cílios/metabolismo , Receptor de Fator de Crescimento Neural/metabolismo , Receptores de Detecção de Cálcio/metabolismo , Doença de Alzheimer/metabolismo , Peptídeos beta-Amiloides/metabolismo , Animais , Morte Celular , Giro Denteado/fisiopatologia , Hipocampo/metabolismo , Hipocampo/fisiopatologia , Humanos , Neurogênese , Neurônios/citologia , Neurônios/metabolismo , Transdução de Sinais
17.
J Neurol Sci ; 322(1-2): 137-40, 2012 Nov 15.
Artigo em Inglês | MEDLINE | ID: mdl-22841885

RESUMO

Alzheimer's disease (AD) is the most common human neurodegenerative ailment, the most prevalent (>95%) late-onset type of which has a still uncertain etiology. The progressive decline of cognitive functions, dementia, and physical disabilities of AD is caused by synaptic losses that progressively disconnect key neuronal networks in crucial brain areas, like the hippocampus and temporoparietal cortex, and critically impair language, sensory processing, memory, and conscious thought. AD's two main hallmarks are fibrillar amyloid-ß (fAß) plaques in extracellular spaces and intracellular accumulation of fAß peptides and neurofibrillary tangles (NFTs). It is still undecided whether either or both these AD hallmarks cause or result from the disease. Recently, the dysregulation of calcium homeostasis has been advanced as a novel cause of AD. In this case, a suitable candidate of AD driver would be the Aß peptides-binding/activated calcium-sensing receptor (CaSR), whose intracellular signalling is triggered by Aß peptides. In this review, we briefly discuss CaSR's roles in normal adult human astrocytes (NAHAs) and their possible impacts on AD.


Assuntos
Doença de Alzheimer/patologia , Encéfalo/metabolismo , Receptores de Detecção de Cálcio/metabolismo , Doença de Alzheimer/complicações , Astrócitos/metabolismo , Astrócitos/patologia , Transtornos Cognitivos/etiologia , Humanos , Emaranhados Neurofibrilares/patologia , Placa Amiloide
18.
Oncol Rep ; 23(4): 887-92, 2010 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-20204270

RESUMO

Perylenequinones that generate reactive oxygen species (ROS) when illuminated with visible light have been recommended as photodynamic chemotherapeutic agents. One of these is calphostin C (CalC), the action of the photo-activated derivative of which, CalCphiE, has been ascribed to its ability to selectively and irreversibly inhibit protein kinase Cs (PKCs). But recent results of experiments with neoplastic rat fibroblasts and human breast and uterine cervix cancer cells have revealed that the action of CalCphiE involves more than PKC inhibition. Besides suppressing PKC activity, CalCphiE rapidly causes endoplasmic reticulum (ER) stress in breast cancer cells and the selective complete oxidation and proteasomal destruction of the functionally essential nuclear envelope protein lamin B1, in human cervical carcinoma (HCC) cells and neoplastic rat fibroblasts. When these lamin B1-lacking cells are placed in the dark, cytoplasmic membrane-linked PKC activities suddenly rebound and apoptogenesis is initiated as indicated by the immediate release of cytochrome c from mitochondria and later on the activation of caspases. Hence, CalCphiE is a photodynamic cytocidal agent attacking multiple targets in cancer cells and it would be worth determining, even for their best applicative use, whether other perylenequinones also share the so far unexpectedly complex deadly properties of the CalCphiE.


Assuntos
Antibióticos Antineoplásicos/farmacologia , Apoptose/efeitos dos fármacos , Lamina Tipo B/efeitos dos fármacos , Naftalenos/farmacologia , Animais , Núcleo Celular/efeitos dos fármacos , Humanos , Fotoquimioterapia/métodos
19.
Cancer Lett ; 275(1): 9-16, 2009 Mar 08.
Artigo em Inglês | MEDLINE | ID: mdl-18725175

RESUMO

There is much evidence that dietary Ca(2+) loading reduces colon cell proliferation and carcinogenesis in humans and rodents, but during carcinogenesis it becomes ineffective or even tumor-promoting. We are beginning to see how Ca(2+) balances the continuous massive cell production in colon crypts by driving the terminal differentiation and eventually the apoptosis of the cells mainly on the mucosal surface, and how this Ca(2+) control is lost during colon carcinogenesis. The rapid proliferation of the transit-amplifying (TA) progeny of the colon stem cells is driven by the so-called "Wnt" signaling mechanism, which involves the stimulation of proliferogenic genes such as those for c-Myc and cyclin D1 and the silencing of the gene for the cell cycle-stopping p21(Cip1/WAF1) protein by nuclear beta-catenin*Tcf-4 complexes. TA cells avoid mitotic damage and premature apoptosis by expressing the protein survivin. It appears that TA cell cycling stops and terminal differentiation starts when the cells reach a higher level in the crypt where there is enough lumenal Ca(2+) to stimulate the expression and activation of CaSRs (Ca(2+)-sensing receptors), the signals from which stimulate the expression of E-cadherin. Along with this, the APC (adenomatous polyposis coli) protein appears and some of it enters the nucleus. There it makes the TA cells susceptible to the eventual apoptotic balancing by stopping survivin expression and the beta-catenin*Tcf-4 complex from driving further cell cycling by releasing beta-catenin from the nucleus, and delivering it to cytoplasmic APC*axin*GSK-3beta complexes for ultimate proteasomal destruction. Cytoplasmic beta-catenin is then prevented from returning to the nucleus by either being intercepted and destroyed by APC*axin*GSK-3beta complexes or locked by the emerging E-cadherin into membrane adherens junctions which tie the cell into the sheet of proliferatively shut-down cells with APC-dependent cytoskeletons moving to the mouth of the crypt and onto the flat mucosal surface. A common first step in sporadic colon carcinogenesis is the loss of functional APC which disorients upwardly directed migration and causes the retention of nuclear beta-catenin and proliferogenic beta-catenin*Tcf-4 complexes as well as genomic instability. Eventually the balance between cell proliferation and terminal differentiation and death is radically tipped in favour of proliferation by the appearance of apoptosis-resistant, survivin-expressing clones of Ca(2+)-insensitive cells which are locked into the proliferative, mutation-prone mode because of CaSR-disabling gene mutations which prevent the stimulation of E-cadherin expression and terminal differentiation.


Assuntos
Cálcio/metabolismo , Neoplasias do Colo/metabolismo , Receptores de Detecção de Cálcio/metabolismo , Animais , Apoptose , Fatores de Transcrição de Zíper de Leucina e Hélice-Alça-Hélix Básicos , Núcleo Celular/metabolismo , Proliferação de Células , Ciclina D1/metabolismo , Citoplasma/metabolismo , Proteínas de Ligação a DNA/metabolismo , Quinase 3 da Glicogênio Sintase/metabolismo , Glicogênio Sintase Quinase 3 beta , Humanos , Proteínas Inibidoras de Apoptose , Proteínas Associadas aos Microtúbulos/metabolismo , Proteínas Proto-Oncogênicas c-myc/metabolismo , Survivina , Fator de Transcrição 4 , Fatores de Transcrição/metabolismo , beta Catenina/metabolismo
20.
Cell Signal ; 21(9): 1351-5, 2009 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-19249355

RESUMO

Considerable attention has recently been focused on the postnatal persistence of neurogenesis in the dentate gyrus of the hippocampus and the roles of signals from the primary cilium in the different functions of an increasing number of tissues and their malfunctions. Here we summarize the evidence that ties sonic hedgehog-triggered proliferogenic signaling from the primary cilia on granule cell progenitors in the adult dentate subgranular zone to maintain a pool of new "blank slate" dentate granule cells. These can be recruited to bundle and encode novel inputs flowing from various regions of the brain into the dentate gyrus via the entorhinal cortex without altering and erasing the synaptic patterns from previous inputs inscribed on older granule cells.


Assuntos
Giro Denteado/fisiologia , Memória/fisiologia , Neurogênese , Neurônios/fisiologia , Adulto , Animais , Cílios/fisiologia , Giro Denteado/citologia , Humanos , Camundongos , Ratos , Transdução de Sinais
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA