Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 22
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
PLoS One ; 13(9): e0204536, 2018.
Artigo em Inglês | MEDLINE | ID: mdl-30226905

RESUMO

[This corrects the article DOI: 10.1371/journal.pone.0167573.].

2.
J Neurosci ; 37(2): 413-421, 2017 01 11.
Artigo em Inglês | MEDLINE | ID: mdl-28077719

RESUMO

Canavan disease is a leukodystrophy caused by aspartoacylase (ASPA) deficiency. The lack of functional ASPA, an enzyme enriched in oligodendroglia that cleaves N-acetyl-l-aspartate (NAA) to acetate and l-aspartic acid, elevates brain NAA and causes "spongiform" vacuolation of superficial brain white matter and neighboring gray matter. In children with Canavan disease, neuroimaging shows early-onset dysmyelination and progressive brain atrophy. Neuron loss has been documented at autopsy in some cases. Prior studies have shown that mice homozygous for the Aspa nonsense mutation Nur7 also develop brain vacuolation. We now report that numbers of cerebral cortical and cerebellar neurons are decreased and that cerebral cortex progressively thins in AspaNur7/Nur7 mice. This neuronal pathology is prevented by constitutive disruption of Nat8l, which encodes the neuronal NAA-synthetic enzyme N-acetyltransferase-8-like. SIGNIFICANCE STATEMENT: This is the first demonstration of cortical and cerebellar neuron depletion and progressive cerebral cortical thinning in an animal model of Canavan disease. Genetic suppression of N-acetyl-l-aspartate (NAA) synthesis, previously shown to block brain vacuolation in aspartoacylase-deficient mice, also prevents neuron loss and cerebral cortical atrophy in these mice. These results suggest that lowering the concentration of NAA in the brains of children with Canavan disease would prevent or slow progression of neurological deficits.


Assuntos
Ácido Aspártico/análogos & derivados , Doença de Canavan/metabolismo , Modelos Animais de Doenças , Neurônios/metabolismo , Animais , Ácido Aspártico/biossíntese , Ácido Aspártico/deficiência , Ácido Aspártico/genética , Doença de Canavan/genética , Doença de Canavan/patologia , Feminino , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Neurônios/patologia
3.
PLoS One ; 11(12): e0167573, 2016.
Artigo em Inglês | MEDLINE | ID: mdl-27907123

RESUMO

Charcot-Marie-Tooth disease type 2A (CMT2A), the most common axonal form of hereditary sensory motor neuropathy, is caused by mutations of mitofusin-2 (MFN2). Mitofusin-2 is a GTPase required for fusion of mitochondrial outer membranes, repair of damaged mitochondria, efficient mitochondrial energetics, regulation of mitochondrial-endoplasmic reticulum calcium coupling and axonal transport of mitochondria. We knocked T105M MFN2 preceded by a loxP-flanked STOP sequence into the mouse Rosa26 locus to permit cell type-specific expression of this pathogenic allele. Crossing these mice with nestin-Cre transgenic mice elicited T105M MFN2 expression in neuroectoderm, and resulted in diminished numbers of mitochondria in peripheral nerve axons, an alteration in skeletal muscle fiber type distribution, and a gait abnormality.


Assuntos
Alelos , Doença de Charcot-Marie-Tooth/genética , GTP Fosfo-Hidrolases/genética , Hemizigoto , Fibras Musculares Esqueléticas/metabolismo , Nervos Periféricos/metabolismo , Substituição de Aminoácidos , Animais , Transporte Axonal , Doença de Charcot-Marie-Tooth/metabolismo , Doença de Charcot-Marie-Tooth/patologia , Modelos Animais de Doenças , Embrião de Mamíferos , GTP Fosfo-Hidrolases/deficiência , Marcha , Regulação da Expressão Gênica , Membro Posterior/patologia , Humanos , Integrases/genética , Integrases/metabolismo , Camundongos , Camundongos Transgênicos , Mitocôndrias/metabolismo , Mitocôndrias/patologia , Fibras Musculares Esqueléticas/patologia , Nestina/genética , Nestina/metabolismo , Placa Neural/metabolismo , Placa Neural/patologia , Nervos Periféricos/patologia , Fenótipo , RNA não Traduzido/genética , RNA não Traduzido/metabolismo
4.
J Neuroimmunol ; 290: 36-46, 2016 Jan 15.
Artigo em Inglês | MEDLINE | ID: mdl-26711567

RESUMO

Studies in multiple sclerosis and its animal model experimental autoimmune encephalomyelitis (EAE) suggest that peripheral monocyte-derived cells (MDCs) are instrumental for disease initiation. MDCs, however, are plastic, and may exert various functions once in the central nervous system (CNS) for prolonged periods. Furthermore, the long-term effect of MDC depletion on continuing axon loss is not known. We show that long-lasting depletion of MDCs, after onset of EAE clinical deficits, is accompanied by decreased CNS infiltration by pathogenic T lymphocytes. Although this treatment does not reverse clinical disease, it prevents worsening of neurological deficits and long-term axonal loss.


Assuntos
Axônios/metabolismo , Encefalomielite Autoimune Experimental/metabolismo , Encefalomielite Autoimune Experimental/terapia , Monócitos/metabolismo , Animais , Axônios/imunologia , Encefalomielite Autoimune Experimental/imunologia , Leucócitos Mononucleares/imunologia , Leucócitos Mononucleares/metabolismo , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Monócitos/imunologia
5.
Ann Neurol ; 77(5): 884-8, 2015 May.
Artigo em Inglês | MEDLINE | ID: mdl-25712859

RESUMO

Canavan disease is caused by inactivating ASPA (aspartoacylase) mutations that prevent cleavage of N-acetyl-L-aspartate (NAA), resulting in marked elevations in central nervous system (CNS) NAA and progressively worsening leukodystrophy. We now report that ablating NAA synthesis by constitutive genetic disruption of Nat8l (N-acetyltransferase-8 like) permits normal CNS myelination and prevents leukodystrophy in a murine Canavan disease model.


Assuntos
Ácido Aspártico/análogos & derivados , Doença de Canavan/metabolismo , Doença de Canavan/prevenção & controle , Modelos Animais de Doenças , Animais , Ácido Aspártico/deficiência , Ácido Aspártico/genética , Ácido Aspártico/metabolismo , Doença de Canavan/genética , Feminino , Masculino , Camundongos , Camundongos Knockout
6.
J Neurosci ; 34(24): 8175-85, 2014 Jun 11.
Artigo em Inglês | MEDLINE | ID: mdl-24920622

RESUMO

Current multiple sclerosis (MS) therapies only partially prevent chronically worsening neurological deficits, which are largely attributable to progressive loss of CNS axons. Prior studies of experimental autoimmune encephalomyelitis (EAE) induced in C57BL/6 mice by immunization with myelin oligodendrocyte glycoprotein peptide 35-55 (MOG peptide), a model of MS, documented continued axon loss for months after acute CNS inflammatory infiltrates had subsided, and massive astroglial induction of CCL2 (MCP-1), a chemokine for CCR2(+) monocytes. We now report that conditional deletion of astroglial CCL2 significantly decreases CNS accumulation of classically activated (M1) monocyte-derived macrophages and microglial expression of M1 markers during the initial CNS inflammatory phase of MOG peptide EAE, reduces the acute and long-term severity of clinical deficits and slows the progression of spinal cord axon loss. In addition, lack of astroglial-derived CCL2 results in increased accumulation of Th17 cells within the CNS in these mice, but also in greater confinement of CD4(+) lymphocytes to CNS perivascular spaces. These findings suggest that therapies designed to inhibit astroglial CCL2-driven trafficking of monocyte-derived macrophages to the CNS during acute MS exacerbations have the potential to significantly reduce CNS axon loss and slow progression of neurological deficits.


Assuntos
Astrócitos/metabolismo , Axônios/patologia , Sistema Nervoso Central/patologia , Quimiocina CCL2/metabolismo , Encefalomielite Autoimune Experimental/patologia , Macrófagos/metabolismo , Análise de Variância , Animais , Axônios/ultraestrutura , Proteínas de Bactérias/genética , Sistema Nervoso Central/ultraestrutura , Quimiocina CCL2/genética , Citometria de Fluxo , Proteína Glial Fibrilar Ácida/genética , Proteína Glial Fibrilar Ácida/metabolismo , Proteínas Luminescentes/genética , Macrófagos/imunologia , Macrófagos/ultraestrutura , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Microscopia Eletrônica de Transmissão , Glicoproteína Mielina-Oligodendrócito/toxicidade , Fragmentos de Peptídeos/toxicidade , Proteínas/genética
7.
J Neuroinflammation ; 11: 105, 2014 Jun 12.
Artigo em Inglês | MEDLINE | ID: mdl-24924222

RESUMO

Multiple sclerosis (MS) is characterized by central nervous system (CNS) inflammation, demyelination, and axonal degeneration. CXCL10 (IP-10), a chemokine for CXCR3+ T cells, is known to regulate T cell differentiation and migration in the periphery, but effects of CXCL10 produced endogenously in the CNS on immune cell trafficking are unknown. We created floxed cxcl10 mice and crossed them with mice carrying an astrocyte-specific Cre transgene (mGFAPcre) to ablate astroglial CXCL10 synthesis. These mice, and littermate controls, were immunized with myelin oligodendrocyte glycoprotein peptide 35-55 (MOG peptide) to induce experimental autoimmune encephalomyelitis (EAE). In comparison to the control mice, spinal cord CXCL10 mRNA and protein were sharply diminished in the mGFAPcre/CXCL10fl/fl EAE mice, confirming that astroglia are chiefly responsible for EAE-induced CNS CXCL10 synthesis. Astroglial CXCL10 deletion did not significantly alter the overall composition of CD4+ lymphocytes and CD11b+ cells in the acutely inflamed CNS, but did diminish accumulation of CD4+ lymphocytes in the spinal cord perivascular spaces. Furthermore, IBA1+ microglia/macrophage accumulation within the lesions was not affected by CXCL10 deletion. Clinical deficits were milder and acute demyelination was substantially reduced in the astroglial CXCL10-deleted EAE mice, but long-term axon loss was equally severe in the two groups. We concluded that astroglial CXCL10 enhances spinal cord perivascular CD4+ lymphocyte accumulation and acute spinal cord demyelination in MOG peptide EAE, but does not play an important role in progressive axon loss in this MS model.


Assuntos
Astrócitos/metabolismo , Axônios/patologia , Quimiocina CXCL10/deficiência , Encefalomielite Autoimune Experimental/patologia , Animais , Proteínas de Ligação ao Cálcio/metabolismo , Sistema Nervoso Central/patologia , Quimiocina CXCL10/genética , Modelos Animais de Doenças , Progressão da Doença , Encefalomielite Autoimune Experimental/induzido quimicamente , Proteína Glial Fibrilar Ácida/metabolismo , Leucócitos/metabolismo , Linfonodos/patologia , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Proteínas dos Microfilamentos/metabolismo , Proteína Básica da Mielina/metabolismo , Glicoproteína Mielina-Oligodendrócito/toxicidade , Fragmentos de Peptídeos/toxicidade , Medula Espinal/patologia , Baço/patologia , Fatores de Tempo
8.
J Neuropathol Exp Neurol ; 73(4): 335-44, 2014 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-24607968

RESUMO

We provide evidence of cortical neuronopathy in myelin oligodendrocyte glycoprotein peptide-induced experimental autoimmune encephalomyelitis, an established model of chronic multiple sclerosis. To investigate phenotypic perturbations in neurons in this model, we used apoptotic markers and immunohistochemistry with antibodies to NeuN and other surrogate markers known to be expressed by adult pyramidal Layer V somas, including annexin V, encephalopsin, and Emx1. We found no consistent evidence of chronic loss of Layer V neurons but detected both reversible and chronic decreases in the expression of these markers in conjunction with evidence of cortical demyelination and presynaptic loss. These phenotypic perturbations were present in, but not restricted to, the neocortical Layer V. We also investigated inflammatory responses in the cortex and subcortical white matter of the corpus callosum and spinal dorsal funiculus and found that those in the cortex and corpus callosum were delayed compared with those in the spinal cord. Inflammatory infiltrates initially included T cells, neutrophils, and Iba1-positive microglia/macrophages in the corpus callosum, whereas only Iba1-positive cells were present in the cortex. These data indicate that we have identified a new temporal pattern of subtle phenotypic perturbations in neocortical neurons in this chronic multiple sclerosis model.


Assuntos
Neurônios Motores/patologia , Esclerose Múltipla/patologia , Neocórtex/patologia , Animais , Caspase 3/metabolismo , Morte Celular/efeitos dos fármacos , Modelos Animais de Doenças , Encefalite/etiologia , Adjuvante de Freund/toxicidade , Humanos , Marcação In Situ das Extremidades Cortadas , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Neurônios Motores/efeitos dos fármacos , Esclerose Múltipla/induzido quimicamente , Esclerose Múltipla/complicações , Proteína Básica da Mielina/metabolismo , Glicoproteína Mielina-Oligodendrócito/toxicidade , Neocórtex/efeitos dos fármacos , Fragmentos de Peptídeos/toxicidade , Fosfopiruvato Hidratase/metabolismo , Sinaptofisina/metabolismo , Fatores de Tempo
9.
J Neurosci ; 32(2): 639-45, 2012 Jan 11.
Artigo em Inglês | MEDLINE | ID: mdl-22238099

RESUMO

Pharmacological studies have suggested that oligodendroglial NMDA glutamate receptors (NMDARs) mediate white matter injury in a variety of CNS diseases, including multiple sclerosis (MS). We tested this hypothesis in experimental autoimmune encephalomyelitis (EAE), a model of human MS, by timed conditional disruption of oligodendroglial NR1, an essential subunit of functional NMDARs, using an inducible proteolipid protein (Plp) promoter-driven Cre-loxP recombination system. We found that selective ablation of oligodendroglial NR1 did not alter the clinical severity of EAE elicited in C57BL/6 mice by immunization with myelin oligodendrocyte glycoprotein peptide 35-55 (MOG-peptide), nor were there significant differences between the oligodendroglial NR1 KO and non-KO mice in numbers of axons lost in spinal cord dorsal funiculi or severity of spinal cord demyelination. Similarly, constitutive deletion of NR3A, a modulatory subunit of oligodendroglial NMDARs, did not alter the course of MOG-peptide EAE. Furthermore, conditional and constitutive ablation of NR1 in neonatal oligodendrocyte progenitor cells did not interrupt their normal maturation and differentiation. Collectively, our data suggest that oligodendroglial lineage NMDARs are neither required for timely postnatal development of the oligodendroglial lineage, nor significant participants in the pathophysiology of MOG-peptide EAE.


Assuntos
Linhagem da Célula/fisiologia , Encefalomielite Autoimune Experimental/metabolismo , Encefalomielite Autoimune Experimental/patologia , Oligodendroglia/metabolismo , Receptores de N-Metil-D-Aspartato/deficiência , Animais , Animais Recém-Nascidos , Modelos Animais de Doenças , Suscetibilidade a Doenças , Encefalomielite Autoimune Experimental/fisiopatologia , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Oligodendroglia/patologia , Oligodendroglia/fisiologia , Receptores de N-Metil-D-Aspartato/genética , Receptores de N-Metil-D-Aspartato/metabolismo
10.
J Neurosci ; 31(33): 11914-28, 2011 Aug 17.
Artigo em Inglês | MEDLINE | ID: mdl-21849552

RESUMO

Accumulations of hypertrophic, intensely glial fibrillary acidic protein-positive (GFAP(+)) astroglia, which also express immunoreactive nestin and vimentin, are prominent features of multiple sclerosis lesions. The issues of the cellular origin of hypertrophic GFAP(+)/vimentin(+)/nestin(+) "reactive" astroglia and also the plasticities and lineage relationships among three macroglial progenitor populations-oligodendrocyte progenitor cells (OPCs), astrocytes and ependymal cells-during multiple sclerosis and other CNS diseases remain controversial. We used genetic fate-mappings with a battery of inducible Cre drivers (Olig2-Cre-ER(T2), GFAP-Cre-ER(T2), FoxJ1-Cre-ER(T2) and Nestin-Cre-ER(T2)) to explore these issues in adult mice with myelin oligodendrocyte glycoprotein peptide-induced experimental autoimmune encephalomyelitis (EAE). The proliferative rate of spinal cord OPCs rose fivefold above control levels during EAE, and numbers of oligodendroglia increased as well, but astrogenesis from OPCs was rare. Spinal cord ependymal cells, previously reported to be multipotent, did not augment their low proliferative rate, nor give rise to astroglia or OPCs. Instead, the hypertrophic, vimentin(+)/nestin(+), reactive astroglia that accumulated in spinal cord in this multiple sclerosis model were derived by proliferation and phenotypic transformation of fibrous astroglia in white matter, and solely by phenotypic transformation of protoplasmic astroglia in gray matter. This comprehensive analysis of macroglial plasticity in EAE helps to clarify the origins of astrogliosis in CNS inflammatory demyelinative disorders.


Assuntos
Astrócitos/patologia , Encefalomielite Autoimune Experimental/patologia , Plasticidade Neuronal/fisiologia , Oligodendroglia/patologia , Animais , Astrócitos/fisiologia , Feminino , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Oligodendroglia/fisiologia , Medula Espinal/citologia , Medula Espinal/fisiologia
11.
J Neurosci ; 30(36): 12036-49, 2010 Sep 08.
Artigo em Inglês | MEDLINE | ID: mdl-20826667

RESUMO

Previous studies have shown that oligodendroglial progenitor cells (OPCs) can give rise to neurons in vitro and in perinatal cerebral cortex in vivo. We now report that OPCs in adult murine piriform cortex express low levels of doublecortin, a marker for migratory and immature neurons. Additionally, these OPCs express Sox2, a neural stem cell marker, and Pax6, a transcription factor characteristic of progenitors for cortical glutamatergic neurons. Genetic fate-mapping by means of an inducible Cre-LoxP recombination system proved that these OPCs differentiate into pyramidal glutamatergic neurons in piriform cortex. Several lines of evidence indicated that these newly formed neurons became functionally integrated into the cortical neuronal network. Our data suggest that NG2(+)/PDGFRα(+) proteolipid protein promoter-expressing progenitors generate pyramidal glutamatergic neurons within normal adult piriform cortex.


Assuntos
Células-Tronco Adultas/fisiologia , Diferenciação Celular/fisiologia , Córtex Cerebral/citologia , Oligodendroglia/fisiologia , Células Piramidais/fisiologia , Células-Tronco Adultas/efeitos dos fármacos , Animais , Antígenos/metabolismo , Antineoplásicos Hormonais/farmacologia , Bromodesoxiuridina/metabolismo , Contagem de Células/métodos , Diferenciação Celular/efeitos dos fármacos , Diferenciação Celular/genética , Proteínas do Domínio Duplacortina , Esquema de Medicação , Proteínas do Olho/metabolismo , Regulação da Expressão Gênica/efeitos dos fármacos , Regulação da Expressão Gênica/genética , Proteínas de Fluorescência Verde/genética , Proteínas de Homeodomínio/metabolismo , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Proteínas Associadas aos Microtúbulos/metabolismo , Proteína Proteolipídica de Mielina/genética , Proteínas do Tecido Nervoso/metabolismo , Neurônios/fisiologia , Neuropeptídeos/metabolismo , Fator de Transcrição PAX6 , Fatores de Transcrição Box Pareados/metabolismo , Proteoglicanas/metabolismo , Receptor alfa de Fator de Crescimento Derivado de Plaquetas/metabolismo , Receptores de N-Metil-D-Aspartato/metabolismo , Proteínas Repressoras/metabolismo , Fatores de Transcrição SOXB1/metabolismo , Tamoxifeno/farmacologia , Fatores de Tempo
12.
J Neurosci ; 29(47): 14965-79, 2009 Nov 25.
Artigo em Inglês | MEDLINE | ID: mdl-19940192

RESUMO

Axonal loss is the principal cause of chronic disability in multiple sclerosis and experimental autoimmune encephalomyelitis (EAE). In C57BL/6 mice with EAE induced by immunization with myelin oligodendrocyte glycoprotein peptide 35-55, the first evidences of axonal damage in spinal cord were in acute subpial and perivascular foci of infiltrating neutrophils and lymphocytes and included intra-axonal accumulations of the endovesicular Toll-like receptor TLR8, and the inflammasome protein NAcht leucine-rich repeat protein 1 (NALP1). Later in the course of this illness, focal inflammatory infiltrates disappeared from the spinal cord, but there was persistent activation of spinal cord innate immunity and progressive, bilaterally symmetric loss of small-diameter corticospinal tract axons. These results support the hypothesis that both contact-dependent and paracrine interactions of systemic inflammatory cells with axons and an innate immune-mediated neurodegenerative process contribute to axonal loss in this multiple sclerosis model.


Assuntos
Axônios/patologia , Encefalomielite Autoimune Experimental/patologia , Medula Espinal/patologia , Degeneração Walleriana/patologia , Proteínas Adaptadoras de Transdução de Sinal/metabolismo , Animais , Proteínas Reguladoras de Apoptose/metabolismo , Axônios/metabolismo , Quimiotaxia de Leucócito/imunologia , Modelos Animais de Doenças , Progressão da Doença , Encefalomielite Autoimune Experimental/induzido quimicamente , Encefalomielite Autoimune Experimental/fisiopatologia , Imunidade Inata/imunologia , Leucócitos/patologia , Linfócitos/patologia , Camundongos , Camundongos Endogâmicos C57BL , Esclerose Múltipla/imunologia , Esclerose Múltipla/patologia , Esclerose Múltipla/fisiopatologia , Neutrófilos/patologia , Tratos Piramidais/imunologia , Tratos Piramidais/patologia , Tratos Piramidais/fisiopatologia , Medula Espinal/imunologia , Medula Espinal/fisiopatologia , Receptor 8 Toll-Like/metabolismo , Degeneração Walleriana/imunologia , Degeneração Walleriana/fisiopatologia
13.
J Nucl Med ; 48(8): 1338-47, 2007 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-17631545

RESUMO

UNLABELLED: Despite their large size, antibodies (Abs) are suitable carriers to deliver systemic radiotherapy, often molecular image-based, for lymphoma and leukemia. Lym-1 Ab has proven to be an effective radioisotope carrier, even in small amounts, for targeting human leukocyte antigen DR (HLA-DR), a surface membrane protein overexpressed on B-cell lymphoma. Pairs of molecules (referred to as ligands), shown by computational and experimental methods to bind to each of 2 sites within the Lym-1 epitopic region, have been linked to generate small (<2 kDa) molecules (referred to as selective high-affinity ligands [SHALs]) to mimic the targeting properties of Lym-1 Ab. METHODS: A lysine-polyethylene glycol (PEG) backbone was used to synthetically link 2 of the following ligands: deoxycholate, 5-leuenkephalin, triiodothyronine, thyronine, dabsyl-L-valine, and N-benzoyl-L-arginyl-4-amino-benzoic acid to generate a series of 13 bidentate SHALs with a biotin or 1,4,7,10-tetraazacyclododecane-N,N',N'',N'''-tetraacetic acid (DOTA) chelate attached to the linker. These SHALs have been assessed for their selectivity in binding to HLA-DR10-expressing cells and for their pharmacokinetics and tissue biodistribution in mice. Biotinylated versions of these SHALs discriminated cell lines positive for HLA-DR10 expression with near-nanomolar affinity. The DOTA versions of 4 SHALs were labeled with (111)In for pharmacokinetic studies in mice with HLA-DR10-expressing malignant Raji xenografts. RESULTS: The bidentate, biotinylated, and DOTA-SHALs were synthesized in high-purity, multimilligram amounts. Mean radiochemical and product yields and purities were 90%, 75%, and 90% at mean specific activities of 3.9 MBq/microg (105 microCi/microg) for the (111)In-labeled SHALs. As expected, rapid blood clearance and tumor targeting were observed. The pharmacokinetics of the SHALs was influenced by the component ligands. Biliary clearance, kidney localization, and serum receptor binding contributed to less favorable tumor targeting. CONCLUSION: A series of SHALs was readily synthesized in multimilligram amounts and showed the expected selective binding in vitro. Better selection of the SHAL components should provide second-generation SHALs with improved properties to fulfill the substantial potential of these novel molecular carriers for targeting.


Assuntos
Anticorpos Monoclonais/metabolismo , Antígenos HLA-DR/imunologia , Linfoma não Hodgkin/diagnóstico por imagem , Compostos Radiofarmacêuticos/farmacocinética , Animais , Anticorpos Monoclonais Murinos , Biotinilação , Feminino , Humanos , Ligantes , Linfoma não Hodgkin/radioterapia , Camundongos , Camundongos Endogâmicos BALB C , Transplante de Neoplasias , Tomografia por Emissão de Pósitrons , Compostos Radiofarmacêuticos/síntese química , Transplante Heterólogo
14.
J Nucl Med ; 48(3): 437-44, 2007 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-17332622

RESUMO

UNLABELLED: Antibody (mAb)-linked iron oxide nanoparticles (bioprobes) provide the opportunity to develop tumor specific thermal therapy (Rx) for metastatic cancer when inductively heated by an externally applied alternating magnetic field (AMF). To evaluate the potential of this Rx, in vivo tumor targeting, efficacy, and predictive radionuclide-based heat dosimetry were studied using (111)In-ChL6 bioprobes (ChL6 is chimeric L6) in a human breast cancer xenograft model. METHODS: Using carbodiimide, (111)In-DOTA-ChL6 (DOTA is dodecanetetraacetic acid) was conjugated to polyethylene glycol-iron oxide-impregnated dextran 20-nm particles and purified as (111)In-bioprobes. (111)In doses of 740-1,110 kBq (20-30 muCi) (2.2 mg of bioprobes) were injected intravenously into mice bearing HBT3477 human breast cancer xenografts. Pharmacokinetic (PK) data were obtained at 1, 2, 3, and 5 d. AMF was delivered 72 h after bioprobe injection at amplitudes of 1,410 (113 kA/m), 1,300 (104 kA/m), and 700 (56 kA/m) oersteds (Oe) at 30%, 60%, and 90% "on" time (duty), respectively, and at 1,050 Oe (84 kA/m) at 50% and 70% duty over the 20-min treatment. Treated and control mice were monitored for 90 d. Tumor total heat dose (THD) from activated tumor bioprobes was calculated for each Rx group using (111)In-bioprobe tumor concentration and premeasured particle heat response to AMF amplitudes. Tumor growth delay was analyzed by Wilcoxon rank sum comparison of time to double, triple, and quintuple tumor volume in each group, and all groups were compared with the controls. RESULTS: Mean tumor concentration of (111)In-bioprobes at 48 h was 14 +/- 2 percentage injected dose per gram; this concentration 24 h before AMF treatment was used to calculate THD. No particle-related toxicity was observed. Toxicity was observed at the highest AMF amplitude-duty combination of 1,300 Oe and 60% over 20 min; 6 of 10 mice died acutely. Tumor growth delay occurred in all of the other groups, correlated with heat dose and, except for the lowest heat dose group, was statistically significant when compared with the untreated group. Electron microscopy showed (111)In-bioprobes on tumor cells and cell death by necrosis at 24 and 48 h after AMF. CONCLUSION: mAb-guided bioprobes (iron oxide nanoparticles) effectively targeted human breast cancer xenografts in mice. THD, calculated using empirically observed (111)In-bioprobe tumor concentration and in vitro nanoparticle heat induction by AMF, correlated with tumor growth delay.


Assuntos
Anticorpos Monoclonais/uso terapêutico , Temperatura Alta/uso terapêutico , Radioisótopos de Índio/uso terapêutico , Magnetismo , Neoplasias Mamárias Experimentais/terapia , Nanopartículas , Animais , Feminino , Humanos , Camundongos , Camundongos Endogâmicos BALB C , Transplante de Neoplasias
15.
Clin Cancer Res ; 11(19 Pt 2): 7087s-7092s, 2005 Oct 01.
Artigo em Inglês | MEDLINE | ID: mdl-16203807

RESUMO

OBJECTIVES: (111)In-chimeric L6 (ChL6) monoclonal antibody (mAb)-linked iron oxide nanoparticle (bioprobes) pharmacokinetics, tumor uptake, and the therapeutic effect of inductively heating these bioprobes by externally applied alternating magnetic field (AMF) were studied in athymic mice bearing human breast cancer HBT 3477 xenografts. Tumor cell radioimmunotargeting of the bioprobes and therapeutic and toxic responses were determined. METHODS: Using 1-ethyl-3-(3-dimethylaminopropyl)-carbodiimide HCl, (111)In-7,10-tetra-azacyclododecane-N, N',N'',N'''-tetraacetic acid-ChL6 was conjugated to the carboxylated polyethylene glycol on dextran-coated iron oxide 20 nm particles, one to two mAbs per nanoparticle. After magnetic purification and sterile filtration, pharmacokinetics, histopathology, and AMF/bioprobe therapy were done using (111)In-ChL6 bioprobe doses (20 ng/2.2 mg ChL6/ bioprobe), i.v. with 50 microg ChL6 in athymic mice bearing HBT 3477; a 153 kHz AMF was given 72 hours postinjection for therapy with amplitudes of 1,300, 1,000, or 700 Oe. Weights, blood counts, and tumor size were monitored and compared with control mice receiving nothing, or AMF or bioprobes alone. RESULTS: (111)In-ChL6 bioprobe binding in vitro to HBT 3477 cells was 50% to 70% of that of (111)In-ChL6. At 48 hours, tumor, lung, kidney, and marrow uptakes of the (111)In-ChL6 bioprobes were not different from that observed in prior studies of (111)In-ChL6. Significant therapeutic responses from AMF/bioprobe therapy were shown with up to eight times longer mean time to quintuple tumor volume with therapy compared with no treatment (P = 0.0013). Toxicity was only seen in the 1,300 Oe AMF cohort, with 4 of 12 immediate deaths and skin erythema. Electron micrographs showed bioprobes on the surfaces of the HBT 3477 cells of excised tumors and tumor necrosis 24 hours after AMF/bioprobe therapy. CONCLUSION: This study shows that mAb-conjugated nanoparticles (bioprobes), when given i.v., escape into the extravascular space and bind to cancer cell membrane antigen, so that bioprobes can be used in concert with externally applied AMF to deliver thermoablative cancer therapy.


Assuntos
Anticorpos Monoclonais/química , Técnicas Biossensoriais , Radioisótopos de Índio/uso terapêutico , Neoplasias/metabolismo , Neoplasias/terapia , Radioimunoterapia/métodos , Animais , Quelantes/farmacologia , Estudos de Coortes , Campos Eletromagnéticos , Compostos Heterocíclicos com 1 Anel/química , Imunoconjugados/metabolismo , Camundongos , Camundongos Nus , Microscopia Eletrônica , Transplante de Neoplasias , Neoplasias/patologia , Polietilenoglicóis/química , Fatores de Tempo , Distribuição Tecidual
16.
Clin Cancer Res ; 11(19 Pt 2): 7158s-7163s, 2005 Oct 01.
Artigo em Inglês | MEDLINE | ID: mdl-16203816

RESUMO

PURPOSE: Paclitaxel synergized radiolabeled monoclonal antibodies, enhancing therapeutic effect in studies in mice with human xenografts. Paclitaxel was also observed to increase tumor uptake in imaging studies of (111)In-DOTA-Gly3Phe-m170 in patients with breast and prostate cancers. Further evaluations of tissue-cumulated activities, therapeutic indices, and pharmacokinetics were done using data for patients with breast and prostate cancer and for mice with human breast cancer xenografts. EXPERIMENTAL DESIGN: In radioimmunotherapy trials, 12 patients with breast or prostate cancer were given two imaging doses (5 mCi each) of (111)In-DOTA-Gly3Phe-m170 1 week apart. Five of these patients were given a single dose of paclitaxel i.v. (75 mg/m2) 2 days after the second dose of (111)In. In a subsequent study, athymic mice with human breast cancer xenografts were given (111)In-DOTA-Gly3Phe-ChL6 alone, or in combination with daily paclitaxel i.p. (300 microg) one or more times. Pharmacokinetics were studied for at least 6 days in patients and 5 days in mice. Cumulated activities were determined for tumors and normal tissues. RESULTS: Tumor-cumulated activity for every patient in the paclitaxel-treated group increased for the second dose of (111)In-DOTA-Gly3Phe-m170. The median ratio of cumulated activities in tumors for imaging dose 2 to those for dose 1 was 1.0 (0.8-1.3) in patients that were not given paclitaxel and 1.3 (1.2-1.4) in patients given paclitaxel. Normal tissue-cumulated activities were not different for the two doses. Mice given paclitaxel 1 day after (111)In-DOTA-Gly3Phe-ChL6 also showed an increase in tumor-cumulated activity, 22.9 (+/- 1.3) versus 19.4 (+/- 3.3) microCi h/g/microCi (P = 0.05). Cumulated activities of normal tissues were similar for all groups of mice. CONCLUSIONS: Paclitaxel given 1 to 2 days after (111)In-DOTA-Gly3Phe-monoclonal antibody increased the tumor-cumulated activity in patients and in mice with epithelial cancers and did not alter cumulated activities in normal tissues.


Assuntos
Anticorpos Monoclonais/uso terapêutico , Antineoplásicos Fitogênicos/farmacologia , Radioisótopos de Índio/uso terapêutico , Neoplasias Epiteliais e Glandulares/terapia , Paclitaxel/farmacologia , Radioimunoterapia/métodos , Animais , Anticorpos Monoclonais/química , Antineoplásicos Fitogênicos/administração & dosagem , Neoplasias da Mama/patologia , Neoplasias da Mama/terapia , Linhagem Celular Tumoral , Terapia Combinada , Feminino , Compostos Heterocíclicos com 1 Anel/farmacologia , Humanos , Imunoglobulina G/química , Masculino , Neoplasias Mamárias Experimentais/patologia , Neoplasias Mamárias Experimentais/terapia , Camundongos , Transplante de Neoplasias , Paclitaxel/administração & dosagem , Neoplasias da Próstata/patologia , Neoplasias da Próstata/terapia , Fatores de Tempo
17.
Cancer Biother Radiopharm ; 20(6): 614-9, 2005 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-16398613

RESUMO

PURPOSE: Human xenografts in athymic mice are frequently used as preclinical models of cancer to investigate the targeting of drugs. In order to distinguish specific from nonspecific targeting of the xenograft, the mice can be implanted with different malignant cell lines. We studied in xenograft success and growth rates after implantation of human lymphoma and breast cancer cells to begin an assessment of the validity of this approach for distinguishing specific from nonspecific targeting. Investigations were undertaken to determine the effect of two different cell-line xenografts, and prior radiation needed for one of the xenografts, on implantation success and growth rates. EXPERIMENTAL DESIGN: Female athymic mice were given 4 Gy of external beam radiation 4 days prior to subcutaneous (s.c.) abdominal implantation of 6 x 10(6) Raji human lymphoma cells. One week later, 3 x 10(6) hamster blood transfusion (HBT) 3477 human breast cancer cells were implanted s.c. in a contralateral abdominal site. Xenografts were evaluated frequently thereafter. Xenograft success and growth rates were compared to those observed in "historical" control groups, wherein only a single xenograft of each type was implanted. RESULTS: Raji xenografts developed from 73.7% of the implantations, and 100% of the HBT 3477 xenografts were successful in the experimental group. The "historical" Raji xenograft success rate was 74.1% (+/-9.3%), and the "historical" HBT 3477 xenografts success rate was 99.0% (+/-1.1%). HBT 3477 xenografts did not affect the growth rate of the Raji xenografts, and the mean doubling time for the experimental Raji xenografts was 6.3 days (+/-4.5 days), compared to the "historical" control group mean of 5.1 days (+/-3.9 days; p = 0.2). Similarly, the growth rates for the HBT 3477 xenografts were not affected by the Raji xenografts and the pre-radiation needed for this model. Mean doubling time for HBT 3477 xenografts in the presence of Raji xenografts was 9.2 days (+/-17.6 days), compared to a doubling time of 1.4 days (+/-15.2 days; p = 0.55 and 0.94 studies 1 and 2, respectively). Mean HBT 3477 xenograft doubling time for the "historical" control group was 4.4 days (+/-6.0 days). CONCLUSIONS: Implantation of Raji xenografts and HBT 3477 breast cancer xenografts in the same mouse did not affect xenograft success or growth rates, even when whole-body radiation of 4 Gy was used to promote Raji xenografts. These observations are not intended to imply an absence of differences in other biological parameters in this sytem or to encourage extrapolation of the conclusions indiscriminately to other preclinical models. Contrarily, our aim was to encourage other investigators to further validate these frequently used approaches.


Assuntos
Transplante de Neoplasias/métodos , Transplante Heterólogo/métodos , Adenocarcinoma , Animais , Neoplasias da Mama , Linfoma de Burkitt , Divisão Celular , Linhagem Celular Tumoral , Feminino , Humanos , Cinética , Camundongos , Camundongos Nus , Transplante de Neoplasias/patologia , Transplante Heterólogo/patologia
18.
Clin Cancer Res ; 9(10 Pt 2): 3865S-72S, 2003 Sep 01.
Artigo em Inglês | MEDLINE | ID: mdl-14506184

RESUMO

PURPOSE: Immunoglobulins are catabolized in the hepatocytes, primarily by cathepsins. The liver becomes the likely dose-limiting tissue for radiometals, like (90)Y, in radioimmunoconjugates (RICs) used for radioimmunotherapy in combination with bone marrow support. To assess whether in vitro cathepsin-degradable peptide linkers between the chelated radiometal and the antibody decreased hepatic radiation dose, cumulated activity was used as a surrogate for radiation dose. EXPERIMENTAL DESIGN: Four different cathepsin-degradable peptides used to link 1,4,7,10-tetraazacyclododecane-N,N',N",N"'-tetraacetic acid-chelated (111)In to two different monoclonal antibodies were studied in athymic mouse models of human breast cancer or lymphoma. Measured concentrations of activity during 5 days were used to reflect pharmacokinetic behavior for normal tissues and tumor. With the use of linear regression to fit a monoexponential decay function, cumulated activities in the liver and xenografts were calculated. RESULTS: The pharmacokinetic behavior of the cathepsin-degradable peptide-linked RICs was similar to that for the 2-iminothiolane (2IT) nondegradable linked RICs except for the liver. The liver cumulated activities of peptide-linked RICs were significantly decreased from those of the corresponding 2IT-linked RICs, varying between reductions of 59 and 68%. Cumulated activities of peptide-linked RICs in the xenografts were as great as those of 2IT RICs, so that the therapeutic indices (tumor: liver cumulated activity ratios) were substantially better for cathepsin-degradable peptide-linked RICs. CONCLUSIONS: Cathepsin-degradable peptides used to link chelated radiometals to antibodies reduce liver radiation dose and improve the therapeutic index for radioimmunotherapy given in combination with bone marrow support.


Assuntos
Catepsinas/química , Imunoconjugados/química , Fígado/efeitos da radiação , Animais , Anticorpos Monoclonais/química , Neoplasias da Mama/patologia , Quelantes/farmacologia , Cromatografia Líquida de Alta Pressão , Compostos Heterocíclicos com 1 Anel/farmacologia , Humanos , Radioisótopos de Índio/farmacocinética , Modelos Lineares , Fígado/efeitos dos fármacos , Fígado/metabolismo , Linfoma/patologia , Camundongos , Camundongos Nus , Modelos Químicos , Transplante de Neoplasias , Peptídeos/química , Radioimunoterapia/métodos , Fatores de Tempo
19.
Blood ; 101(9): 3641-7, 2003 May 01.
Artigo em Inglês | MEDLINE | ID: mdl-12511412

RESUMO

CD22 is a membrane glycophosphoprotein found on nearly all healthy B-lymphocytes and most B-cell lymphomas. Recent in vitro studies have identified several anti-CD22 monoclonal antibodies (mAbs) that block the interaction of CD22 with its ligand. One of these mAbs, HB22.7, has been shown to effectively induce apoptosis in several B-cell lymphoma cell lines. Lymphoma xenograft studies with Raji-xenograft mice were used to assess the toxicity and efficacy of HB22.7 alone and with combined modality immunotherapy (CMIT) with yttrium (90)Y-DOTA-peptide-Lym-1 radioimmunotherapy (RIT). The effect of the sequence of these agents on the combined treatment was assessed by administering HB22.7 24 hours before, simultaneously with, or 24 hours after RIT. Within the groups treated with RIT alone or with RIT and HB22.7 (CMIT), the reduction in tumor volume was the greatest when HB22.7 was administered simultaneously with and 24 hours after RIT, and in the RIT treatment groups, this translated into the greatest overall response and survival, respectively. Overall survival rates at the end of the 84-day CMIT trial were 67% and 50% in the groups treated with HB22.7 simultaneously and 24 hours after RIT, respectively. This compared favorably with the untreated and the RIT alone groups, which had survival rates of 38% and 43% at the end of the trial. Surprisingly, when compared with untreated controls and all other treatment groups, the greatest cure and overall survival rates were observed in the group treated with HB22.7 alone, with 47% cured and 76% surviving at the end of the 84-day trial. RIT clearance was not affected by treatment with HB22.7. When compared with RIT alone, there was no significant additional hematologic (white blood cell, red blood cell, or platelet count) toxicity when HB22.7 was added to RIT. Nonhematologic toxicity (assessed as change in body weight) was also unchanged when HB22.7 was added to RIT. Thus the anti-CD22 ligand-blocking antibody HB22.7 has independent lymphomacidal properties and augments the efficacy of (90)Y-DOTA-peptide-Lym-1 in lymphoma xenografts without significant toxicity.


Assuntos
Anticorpos Monoclonais/uso terapêutico , Antígenos CD/imunologia , Antígenos de Diferenciação de Linfócitos B/imunologia , Antígenos de Neoplasias/imunologia , Antineoplásicos/uso terapêutico , Linfoma de Burkitt/terapia , Moléculas de Adesão Celular , Imunização Passiva , Imunoconjugados/uso terapêutico , Lectinas/imunologia , Radioimunoterapia , Animais , Anticorpos Monoclonais/imunologia , Anticorpos Monoclonais/farmacocinética , Anticorpos Monoclonais/farmacologia , Anticorpos Monoclonais Murinos , Apoptose , Linfoma de Burkitt/radioterapia , Sinergismo Farmacológico , Feminino , Humanos , Imunoconjugados/farmacocinética , Camundongos , Camundongos Nus , Lectina 2 Semelhante a Ig de Ligação ao Ácido Siálico , Ensaios Antitumorais Modelo de Xenoenxerto
20.
Cancer Res ; 62(15): 4263-72, 2002 Aug 01.
Artigo em Inglês | MEDLINE | ID: mdl-12154028

RESUMO

Although metastatic breast cancer is responsive to radioimmunotherapy (RIT), a systemic targeted radiation modality, complete and permanent remissions are not typical with single-modality treatment. Antiangiogenic agents, which target normal, proliferating endothelial cells, have the potential to provide relatively nontoxic continuous inhibition of tumor growth by blocking new blood vessel growth and may synergize with RIT to increase efficacy. This study was designed to determine whether, and how, the cyclic Arg-Gly-Asp peptide Cilengitide (EMD 121974), which targets the alpha(v)beta(3) integrin receptor expressed on neovasculature, could increase systemic RIT efficacy of therapy in a human breast cancer tumor model having mutant p53 and expressing bcl-2. HBT 3477 breast cancer tumor response in nude mice was compared between groups of untreated mice (n = 24), Cilengitide-treated mice (n = 18), RIT (200-260 mu Ci (90)Y-labeled 1,4,7,10-tetraazacyclododecane-N,N',N",N"'-tetraacetic acid (DOTA)-peptide ChL6; n = 46), and combined modality RIT (CMRIT) using RIT and six doses of Cilengitide (250 microg/dose; n = 41). Tumor size, survival, body weight, and blood counts were monitored for efficacy and toxicity of therapy. To clarify the mechanism of synergistic effect, tumors were evaluated at selected time points through 6 days for apoptosis, proliferation, and microvessel density. Cilengitide alone did not alter tumor growth when compared with untreated mice, but CMRIT with Cilengitide increased efficacy of treatment, with the cure rate for mice that received 260 mu Ci RIT increasing from 15 to 53% (P = 0.011). Lower-dose RIT (200 mu Ci) combined with Cilengitide resulted in less increase in cures (36 compared with 25% for RIT alone; P = 0.514). Combined analysis for high- and low-dose groups demonstrated increased efficacy of CMRIT (P = 0.020). Analysis of tumors from CMRIT mice indicated significantly increased apoptosis of tumor and endothelial cells 5 days after RIT compared with tumors from mice given RIT alone. Proliferation was decreased in CMRIT tumors compared with RIT tumors at 6 days (ANOVA, P < 0.05). Microvessel density in tumors from RIT and CMRIT mice was not different. No increased toxicity attributable to Cilengitide was observed based upon pooled blood sample and no statistical increase in mortality. In conclusion, CMRIT, combining Cilengitide and RIT, significantly increased the efficacy of therapy and increased apoptosis compared with single-modality therapy with either agent, in an aggressive, well-studied breast cancer model. The enhanced therapeutic synergy is of particular note, having been achieved without additional toxicity.


Assuntos
Adenocarcinoma/terapia , Inibidores da Angiogênese/farmacologia , Neoplasias da Mama/terapia , Radioimunoterapia , Receptores de Vitronectina/metabolismo , Venenos de Serpentes/farmacologia , Adenocarcinoma/irrigação sanguínea , Adenocarcinoma/tratamento farmacológico , Adenocarcinoma/radioterapia , Animais , Apoptose/efeitos dos fármacos , Apoptose/efeitos da radiação , Neoplasias da Mama/irrigação sanguínea , Neoplasias da Mama/tratamento farmacológico , Neoplasias da Mama/radioterapia , Divisão Celular/efeitos dos fármacos , Divisão Celular/efeitos da radiação , Terapia Combinada , Endotélio Vascular/citologia , Endotélio Vascular/efeitos dos fármacos , Endotélio Vascular/efeitos da radiação , Feminino , Humanos , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Nus , Ensaios Antitumorais Modelo de Xenoenxerto
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...