Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 25
Filtrar
1.
Cell Biol Int ; 48(2): 216-228, 2024 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-38081783

RESUMO

Triple-negative breast cancer (TNBC) is the most aggressive subtype of breast cancer (BC) compared to other BC subtypes in clinical settings. Currently, there are no effective therapeutic strategies for TNBC treatment. Therefore, there is an urgent need to identify suitable biomarkers or therapeutic targets for TNBC patients. Thrombomodulin (TM) plays a role in cancer progression and metastasis in many different cancers. However, the role of TM in TNBC is not yet fully understood. First, silenced-TM in MDA-MB-231 cells caused an increase in proliferative and metastatic activity. In contrast, overexpression of TM in Hs578T cells caused a reduction in proliferation, invasion, and migration rate. Using RNA-seq analysis, we found that Integrin beta 3 (ITGB3) expression may be a downstream target of TM. Furthermore, we found an increase in ITGB3 levels in TM-KD cells by QPCR and western blot analysis but a decrease in ITGB3 levels in TM-overexpressing cells. We found phospho-smad2/3 levels were increased in TM-KD cells but decreased in TM-overexpressing cells. This implies that TM negatively regulates ITGB3 levels through the activation of the smad2/3 pathway. Silencing ITGB3 in TM-KD cells caused a decrease in proliferation and migration. Finally, we found that higher ITGB3 levels were correlated with poor overall survival and relapse-free survival in patients with TNBC. Our results indicated a novel regulatory relationship between TM and ITGB3 in TNBC.


Assuntos
Neoplasias de Mama Triplo Negativas , Humanos , Western Blotting , Linhagem Celular Tumoral , Movimento Celular , Proliferação de Células , Integrina beta3/genética , Trombomodulina/genética , Neoplasias de Mama Triplo Negativas/metabolismo
2.
J Cell Biochem ; 124(6): 907-920, 2023 06.
Artigo em Inglês | MEDLINE | ID: mdl-37183314

RESUMO

Colorectal cancer (CRC) is the primary cause of death from gastrointestinal cancers. Aldehyde dehydrogenase 2 (ALDH2), a crucial mitochondrial enzyme for the oxidative pathway of alcohol metabolism, plays a dual role in cancer progression. In some cancers, it is tumor suppressive; in others, it drives cancer progression. However, whether targeting ALDH2 has any therapeutic implications or prognostic value in CRC is still unclear. Here, we investigated the role of ALDH2 in CRC progression by targeting its enzymatic activity rather than gene expression. We found that inhibiting ALDH2 by CVT-10216 and daidzein significantly decrease migration and stemness properties of both DLD-1 and HCT 116 cells, whereas activating ALDH2 by Alda-1 enhances migration rate. Concomitantly, ALDH2 inhibition by both CVT-10216 and daidzein downregulates the mRNA levels of fibronectin, snail, twist, MMP7, CD44, c-Myc, SOX2, and OCT-4, which are oncogenic in the advanced stage of CRC. Furthermore, Gene Set Enrichment Analysis (GSEA) on ALDH2 co-expressed genes from The Cancer Genome Atlas (TCGA) revealed that MYC target gene sets are upregulated. We found that ALDH2 inhibition decreased the nuclear protein levels of pGSK3ß serine 9 and c-Myc. This suggests that ALDH2 probably targets ß-catenin signaling in CRC cells. Together, our results demonstrate the prognostic value of ALDH2 in CRC as it regulates both CRC stemness and migration. Our findings also propose that the plant-derived isoflavone daidzein could be a potential chemotherapeutic drug targeting ALDH2 in CRC.


Assuntos
Neoplasias Colorretais , beta Catenina , Humanos , Linhagem Celular Tumoral , beta Catenina/genética , beta Catenina/metabolismo , Neoplasias Colorretais/patologia , Transdução de Sinais , Células HCT116 , Regulação Neoplásica da Expressão Gênica , Aldeído-Desidrogenase Mitocondrial/genética , Aldeído-Desidrogenase Mitocondrial/metabolismo
3.
J Transl Med ; 21(1): 505, 2023 07 26.
Artigo em Inglês | MEDLINE | ID: mdl-37496011

RESUMO

BACKGROUND: Colorectal cancer (CRC) is highly prevalent and lethal globally, and its prognosis remains unsatisfactory. Drug resistance is regarded as the main cause of treatment failure leading to tumor recurrence and metastasis. The overexpression of fucosylated epitopes, which are usually modifications of glycoproteins, was reported to occur in various epithelial cancers. However, the effects of treatments that target these antigens in colorectal cancer remain unclear. METHODS: This study investigated the expression of heavily fucosylated glycans (HFGs) in 30 clinical samples from patients with CRC and other normal human tissues. The complement-dependent cytotoxicity was explored in vitro through treatment with anti-HFG monoclonal antibody (mAb) alone or in combination with chemotherapeutic agents. In vivo inhibitory effects were also examined using a xenograft mouse model. RESULTS: Immunohistochemistry staining and western blotting revealed that HFG expression was higher in human colorectal cancer tissues than in normal tissues. In DLD-1 and SW1116 cells, which overexpress fucosylated epitopes, anti-HFG mAb produced observable cytotoxic effects, especially when it was combined with chemotherapeutic agents. The xenograft model also demonstrated that anti-HFG mAb had potent and dose-dependent inhibitory effects on colorectal tumor growth. CONCLUSIONS: As a novel cancer antigen, HFGs are a promising treatment target, and the implementation of anti-HFG mAb treatment for CRC warrants further investigation.


Assuntos
Neoplasias Colorretais , Recidiva Local de Neoplasia , Humanos , Animais , Camundongos , Imuno-Histoquímica , Antígenos , Modelos Animais de Doenças , Epitopos , Polissacarídeos/farmacologia , Neoplasias Colorretais/patologia , Linhagem Celular Tumoral
4.
Int J Mol Sci ; 24(12)2023 Jun 11.
Artigo em Inglês | MEDLINE | ID: mdl-37373155

RESUMO

Colorectal cancer (CRC) is the third most common cancer and a leading cause of cancer-related mortality worldwide. Even with advances in therapy, CRC mortality remains high. Therefore, there is an urgent need to develop effective therapeutics for CRC. PCTAIRE protein kinase 1 (PCTK1) is an atypical member of the cyclin-dependent kinase (CDK) family, and the function of PCTK1 in CRC is poorly understood. In this study, we found that patients with elevated PCTK1 levels had a better overall survival rate in CRC based on the TCGA dataset. Functional analysis also showed that PCTK1 suppressed cancer stemness and cell proliferation by using PCTK1 knockdown (PCTK1-KD) or knockout (PCTK1-KO) and PCTK1 overexpression (PCTK1-over) CRC cell lines. Furthermore, overexpression of PCTK1 decreased xenograft tumor growth and knockout of PCTK1 significantly increased in vivo tumor growth. Moreover, knockout of PCTK1 was observed to increase the resistance of CRC cells to both irinotecan (CPT-11) alone and in combination with 5-fluorouracil (5-FU). Additionally, the fold change of the anti-apoptotic molecules (Bcl-2 and Bcl-xL) and the proapoptotic molecules (Bax, c-PARP, p53, and c-caspase3) was reflected in the chemoresistance of PCTK1-KO CRC cells. PCTK1 signaling in the regulation of cancer progression and chemoresponse was analyzed using RNA sequencing and gene set enrichment analysis (GSEA). Furthermore, PCTK1 and Bone Morphogenetic Protein Receptor Type 1B (BMPR1B) in CRC tumors were negatively correlated in CRC patients from the Timer2.0 and cBioPortal database. We also found that BMPR1B was negatively correlated with PCTK1 in CRC cells, and BMPR1B expression was upregulated in PCTK1-KO cells and xenograft tumor tissues. Finally, BMPR1B-KD partially reversed cell proliferation, cancer stemness, and chemoresistance in PCTK1-KO cells. Moreover, the nuclear translocation of Smad1/5/8, a downstream molecule of BMPR1B, was increased in PCTK1-KO cells. Pharmacological inhibition of Smad1/5/8 also suppressed the malignant progression of CRC. Taken together, our results indicated that PCTK1 suppresses proliferation and cancer stemness and increases the chemoresponse of CRC through the BMPR1B-Smad1/5/8 signaling pathway.


Assuntos
Neoplasias Colorretais , Resistencia a Medicamentos Antineoplásicos , Humanos , Receptores de Proteínas Morfogenéticas Ósseas Tipo I/metabolismo , Linhagem Celular Tumoral , Proliferação de Células , Neoplasias Colorretais/tratamento farmacológico , Neoplasias Colorretais/genética , Neoplasias Colorretais/patologia , Quinases Ciclina-Dependentes/metabolismo , Resistencia a Medicamentos Antineoplásicos/genética , Fluoruracila/farmacologia , Regulação Neoplásica da Expressão Gênica , Transdução de Sinais
5.
Chin J Physiol ; 62(6): 231-240, 2019.
Artigo em Inglês | MEDLINE | ID: mdl-31793458

RESUMO

Lung harbors the growth of primary and secondary tumors. Even though numerous factors regulate the complex signal transduction and cytoskeletal remodeling toward the progression of lung cancer, cyclin-dependent kinase 5 (Cdk5), a previously known kinase in the central nervous system, has raised much attention in the recent years. Patients with aberrant Cdk5 expression also lead to poor survival. Cdk5 has already been employed in various cellular processes which shape the fate of cancer. In lung cancer, Cdk5 mainly regulates tumor suppressor genes, carcinogenesis, cytoskeletal remodeling, and immune checkpoints. Inhibiting Cdk5 by using drugs, siRNA or CRISP-Cas9 system has rendered crucial therapeutic advantage in the combat against lung cancer. Thus, the relation of Cdk5 to lung cancer needs to be addressed in detail. In this review, we will discuss various cellular events modulated by Cdk5 and we will go further into their underlying mechanism in lung cancer.


Assuntos
Neoplasias Pulmonares , Quinase 5 Dependente de Ciclina , Humanos , Transdução de Sinais
6.
Int J Mol Sci ; 20(16)2019 Aug 09.
Artigo em Inglês | MEDLINE | ID: mdl-31395805

RESUMO

Cyclin-dependent kinase 5 (CDK5) is a unique member of the cyclin-dependent kinase family. CDK5 is activated by binding with its regulatory proteins, mainly p35, and its activation is essential in the development of the central nervous system (CNS) and neurodegeneration. Recently, it has been reported that CDK5 plays important roles in regulating various biological and pathological processes, including cancer progression. Concerning prostate cancer, the androgen receptor (AR) is majorly involved in tumorigenesis, while CDK5 can phosphorylate AR and promotes the proliferation of prostate cancer cells. Clinical evidence has also shown that the level of CDK5 is associated with the progression of prostate cancer. Interestingly, inhibition of CDK5 prevents prostate cancer cell growth, while drug-triggered CDK5 hyperactivation leads to apoptosis. The blocking of CDK5 activity by its small interfering RNAs (siRNA) or Roscovitine, a pan-CDK inhibitor, reduces the cellular AR protein level and triggers the death of prostate cancer cells. Thus, CDK5 plays a crucial role in the growth of prostate cancer cells, and AR regulation is one of the important pathways. In this review paper, we summarize the significant studies on CDK5-mediated regulation of prostate cancer cells. We propose that the CDK5-p35 complex might be an outstanding candidate as a diagnostic marker and potential target for prostate cancer treatment in the near future.


Assuntos
Quinase 5 Dependente de Ciclina/metabolismo , Neoplasias da Próstata/patologia , Androgênios/análise , Androgênios/metabolismo , Animais , Apoptose , Carcinogênese/metabolismo , Carcinogênese/patologia , Quinase 5 Dependente de Ciclina/análise , Humanos , Masculino , Próstata/metabolismo , Próstata/patologia , Neoplasias da Próstata/metabolismo , Neoplasias da Próstata/terapia , Receptores Androgênicos/análise , Receptores Androgênicos/metabolismo , Fator de Transcrição STAT3/análise , Fator de Transcrição STAT3/metabolismo
7.
Anticancer Res ; 44(2): 543-553, 2024 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-38307563

RESUMO

BACKGROUND/AIM: Bladder cancer remains a significant global health concern, necessitating a deeper understanding of the molecular mechanisms underlying its progression. Cyclin-Dependent Kinase 5 (CDK5) has recently emerged as a potential player in bladder cancer pathogenesis. This study investigated the involvement of CDK5 in bladder cancer, emphasizing its potential as a therapeutic target. MATERIALS AND METHODS: The expression levels of CDK5 and p35 (CDK5 regulatory protein) and their roles in the tumor grade and malignancy of patient samples were evaluated using western blot analysis and immunohistochemistry. In addition, tumor cancer genome atlas (TCGA) was utilized to evaluate survival rate in patients with bladder cancer. We further confirmed the role of CDK5 with in vitro experiments using western blot analysis, immunocytochemistry, cell culture-based proliferation and migration assays. RESULTS: Higher CDK5 and p35 were associated with a higher tumor grade and poor survival rate in patients with bladder cancer. To confirm the role of CDK5 in vitro, we over-expressed CDK5 in bladder cancer cells. The results showed that the over-expression of CDK5 enhanced bladder cancer cell proliferation and migration. In addition, CDK5 inhibition by a pan-CDK inhibitor, Roscovitine (RV), significantly reduced proliferation of bladder cancer cells. Indeed, the migration and adhesion of bladder cancer cells were inhibited by RV treatment. CONCLUSION: CDK5 might play important roles in bladder cancer progression and be a potential diagnostic and therapeutic target in the near future.


Assuntos
Neoplasias da Bexiga Urinária , Humanos , Proliferação de Células , Quinase 5 Dependente de Ciclina/genética , Quinase 5 Dependente de Ciclina/metabolismo , Roscovitina , Taxa de Sobrevida , Neoplasias da Bexiga Urinária/genética , Neoplasias da Bexiga Urinária/patologia
8.
Chemosphere ; 358: 142124, 2024 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-38677614

RESUMO

Metformin, the most commonly prescribed drug for the treatment of diabetes, is increasingly used during pregnancy to address various disorders such as diabetes, obesity, preeclampsia, and metabolic diseases. However, its impact on neocortex development remains unclear. Here, we investigated the direct effects of metformin on neocortex development, focusing on ERK and p35/CDK5 regulation. Using a pregnant rat model, we found that metformin treatment during pregnancy induces small for gestational age (SGA) and reduces relative cortical thickness in embryos and neonates. Additionally, we discovered that metformin inhibits neural progenitor cell proliferation in the sub-ventricular zone (SVZ)/ventricular zone (VZ) of the developing neocortex, a process possibly mediated by ERK inactivation. Furthermore, metformin induces neuronal apoptosis in the SVZ/VZ area of the developing neocortex. Moreover, metformin retards neuronal migration, cortical lamination, and differentiation, potentially through p35/CDK5 inhibition in the developing neocortex. Remarkably, compensating for p35 through in utero electroporation partially rescues metformin-impaired neuronal migration and development. In summary, our study reveals that metformin disrupts neocortex development by inhibiting neuronal progenitor proliferation, neuronal migration, cortical layering, and cortical neuron maturation, likely via ERK and p35/CDK5 inhibition. Consequently, our findings advocate for caution in metformin usage during pregnancy, given its potential adverse effects on fetal brain development.


Assuntos
Proliferação de Células , Quinase 5 Dependente de Ciclina , Metformina , Neocórtex , Metformina/farmacologia , Animais , Feminino , Gravidez , Neocórtex/efeitos dos fármacos , Quinase 5 Dependente de Ciclina/metabolismo , Ratos , Proliferação de Células/efeitos dos fármacos , Células-Tronco Neurais/efeitos dos fármacos , Células-Tronco Neurais/metabolismo , Sistema de Sinalização das MAP Quinases/efeitos dos fármacos , Neurônios/efeitos dos fármacos , Ratos Sprague-Dawley , Diferenciação Celular/efeitos dos fármacos , Neurogênese/efeitos dos fármacos , Movimento Celular/efeitos dos fármacos , Apoptose/efeitos dos fármacos , Transdução de Sinais/efeitos dos fármacos
9.
Biomedicines ; 11(5)2023 May 07.
Artigo em Inglês | MEDLINE | ID: mdl-37239055

RESUMO

Estrogen and estrogen receptors (ER) play a key role in breast cancer progression, which can be treated with endocrine therapy. Nevertheless, resistance to endocrine therapies is developed over time. The tumor expression of thrombomodulin (TM) is correlated with favorable prognosis in several types of cancer. However, this correlation has not yet been confirmed in ER-positive (ER+) breast cancer. This study aims to evaluate the role of TM in ER+ breast cancer. Firstly, we found that lower TM expression correlates to poor overall survival (OS) and relapse-free survival (RFS) rates in ER+ breast cancer patients through Kaplan-Meier survival analysis (p < 0.05). Silencing TM in MCF7 cells (TM-KD) increased cell proliferation, migration, and invasion ability. Additionally, TM-KD MCF7 cells showed higher sensitivity (IC50 15 µM) to the anti-cancer agent curcumin than the scrambled control cells. Conversely, overexpression of TM (TM-over) in T47D cells leads to decreased cell proliferation, migration, and invasion ability. Furthermore, TM-over T47D cells showed more resistance (IC50 > 40 µM) to the curcumin treatment. The PI staining, DAPI, and tunnel assay also confirmed that the curcumin-induced apoptosis in TM-KD MCF7 cells was higher (90.34%) than in the scrambled control cells (48.54%). Finally, the expressions of drug-resistant genes (ABCC1, LRP1, MRP5, and MDR1) were determined by qPCR. We found that the relative mRNA expression levels of ABCC1, LRP1, and MDR1 genes after curcumin treatment were higher in scrambled control cells than in TM-KD cells. In conclusion, our results demonstrated that TM plays a suppressive role in the progression and metastasis of ER+ breast cancer, and it regulates curcumin sensitivity by interfering with ABCC1, LRP1, and MDR1 gene expression.

10.
J Clin Invest ; 106(5): 689-96, 2000 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-10974022

RESUMO

The genetic defect underlying paroxysmal nocturnal hemoglobinuria (PNH) has been shown to reside in PIGA, a gene that encodes an element required for the first step in glycophosphatidylinositol anchor assembly. Why PIGA-mutated cells are able to expand in PNH marrow, however, is as yet unclear. To address this question, we compared the growth of affected CD59(-)CD34(+) and unaffected CD59(+)CD34(+) cells from patients with that of normal CD59(+)CD34(+) cells in liquid culture. One hundred FACS-sorted cells were added per well into microtiter plates, and after 11 days at 37 degrees C the progeny were counted and were analyzed for their differentiation pattern. We found that CD59(-)CD34(+) cells from PNH patients proliferated to levels approaching those of normal cells, but that CD59(+)CD34(+) cells from the patients gave rise to 20- to 140-fold fewer cells. Prior to sorting, the patients' CD59(-) and CD59(+)CD34(+) cells were equivalent with respect to early differentiation markers, and following culture, the CD45 differentiation patterns were identical to those of control CD34(+) cells. Further analyses of the unsorted CD59(+)CD34(+) population, however, showed elevated levels of Fas receptor. Addition of agonist anti-Fas mAb to cultures reduced the CD59(+)CD34(+) cell yield by up to 78% but had a minimal effect on the CD59(-)CD34(+) cells, whereas antagonist anti-Fas mAb enhanced the yield by up to 250%. These results suggest that expansion of PIGA-mutated cells in PNH marrow is due to a growth defect in nonmutated cells, and that greater susceptibility to apoptosis is one factor involved in the growth impairment.


Assuntos
Células da Medula Óssea/fisiologia , Hemoglobinúria Paroxística/genética , Proteínas de Membrana/genética , Células-Tronco/fisiologia , Receptor fas/metabolismo , Antígenos CD34 , Medula Óssea/cirurgia , Células da Medula Óssea/citologia , Antígenos CD59 , Diferenciação Celular , Separação Celular , Citaferese , Glicosilfosfatidilinositóis/biossíntese , Hemoglobinúria Paroxística/etiologia , Humanos , Fenótipo , Células-Tronco/citologia
11.
Immunology ; 50(1): 11-8, 1983 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-6224741

RESUMO

Experimental antigen-antibody complexes (Ag-Ab) were incubated at 37 degrees with human red blood cells (RBC) suspended in autologous normal serum and the reaction stopped after progressively increasing times. Bound antigen-antibody-complement complexes (Ag-Ab-C) were eluted from C3b receptors and the eluted Ag-Ab-C re-incubated with different blood cell types suspended in serum, or centrifuged (along with unbound Ag-Ab-C found in the serum) through 20-50% sucrose gradients. Ag-Ab-C recovered from C3b receptors shortly after initial binding to RBC bound efficiently to other RBC, polymorphonuclear and mononuclear cells, and sedimented rapidly. Ag-Ab-C simultaneously present in the serum sedimented with a similar velocity. Ag-Ab-C recovered at a subsequent time during RBC interaction bound less well to each blood cell type, and sedimented less rapidly. Decreased amounts of rapidly sedimenting Ag-Ab-C were present in the serum. Ag-Ab-C recovered from C3b receptors at a still later time in the course of RBC interaction bound poorly to each cell type, and sedimented slowly. Increased amounts of slowly sedimenting Ag-Ab-C were found in the serum. These findings indicate that alterations in properties of immune complexes can occur while they are associated with C3b receptors on RBC membrane in solid phase.


Assuntos
Complexo Antígeno-Anticorpo/metabolismo , Membrana Eritrocítica/imunologia , Eritrócitos/imunologia , Humanos , Cinética , Leucócitos/imunologia , Neutrófilos/imunologia , Receptores de Complemento/metabolismo , Receptores de Complemento 3b , Ultracentrifugação
12.
Immunology ; 50(1): 1-9, 1983 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-6411608

RESUMO

Antigen-antibody complexes (Ag-Ab) prepared from 125I-bovine serum albumin (BSA) and guinea-pig anti-BSA were (i) incubated at 37 degrees for 4 min with undiluted normal human serum and autologous red blood cells (RBC) together, or (ii) incubated first at 37 degrees for 30 min with serum diluted optimally for binding and then with RBC. Reactions were stopped by dilution and cooling, and RBC bearing antigen-antibody-complement complexes (Ag-Ab-C) were washed and resuspended in either untreated normal human serum (undiluted or diluted), serum treated with zymosan (SZYM), ethylenediamine tetracetic acid (SEDTA) or ethyleneglycol tetracetic acid-Mg++ (SEGTA), serum heated at 56 degrees for 30 or 120 min (S delta 30 or S delta 120), or buffer alone. The mixtures were placed at 37 degrees and the percentage of Ag-Ab-C dissociated from RBC after progressively increasing times determined. (i) Ag:Ab:C bound to RBC with undiluted serum dissociated more rapidly following resuspension in SZYM, SEDTA, or S delta 30 than following resuspension in untreated serum. Rate of dissociation in SEGTA paralleled that in untreated serum. (ii) Ag-Ab-C bound to RBC with diluted serum dissociated following resuspension in SZYM, SEDTA, or S delta 30, but rebound and dissociated a second time following resuspension in untreated serum or SEGTA. Initial dissociation occurred in less than 1 min in undiluted serum, took place at 0 degrees as well as 37 degrees, and was diminished but detectable in greater than 8- and greater than 64-fold-diluted serum, respectively. Rebinding required 37 degrees, factors B and D and C3, and was maximal at 4-8 min. Subsequent dissociation had similar complement requirements to initial dissociation, but occurred only at 37 degrees and was 90% complete at 15 min. No dissociation of Ag-Ab-C bound to RBC in (i) or in (ii) occurred following resuspension in S delta 120 or in buffer. These findings suggest that after initial binding, release of experimental immune complexes from RBC in whole serum involves concurrent dissociation and alternative pathway-dependent rebinding.


Assuntos
Complexo Antígeno-Anticorpo/metabolismo , Ativação do Complemento , Via Alternativa do Complemento , Eritrócitos/imunologia , Complemento C3/metabolismo , Fator B do Complemento/metabolismo , Fator D do Complemento/metabolismo , Ácido Edético , Ácido Egtázico , Humanos , Cinética , Receptores Imunológicos/metabolismo , Temperatura , Zimosan
13.
Clin Exp Immunol ; 48(3): 715-25, 1982 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-7116690

RESUMO

Antigen-antibody complexes, composed of 125I-BSA and guinea-pig or rabbit antibody, were incubated at 37 degrees C with human blood cells suspended in autologous serum and kinetics of binding analysed. When purified polymorphonuclear (PMN) or mononuclear cells (MNC) were studied, maximum binding was observed within 8 min, and immune complexes (IC) remained associated with cells even after 1 hr. When cells were studied unseparated (in the same amount of serum), maximum binding was observed slightly earlier (within 4 min), but within 15 min most of the IC were found in the serum. Separation of cell types at the time of maximal binding and studies with cell preparations depleted of different elements revealed that binding was principally to red blood cells (RBC). IC recovered in the serum 16 min after addition to unseparated cells bound very slowly to purified PMN or MNC; binding after 30 min was 10-15% of that observed with fresh IC at 8 min. Ultracentrifugal analysis revealed that reduction in binding efficiency correlated with decrease in the size of IC. RBC isolated after binding and release of IC bound newly-formed IC was identical rapidity and capacity as fresh RBC, indicating that receptors were not altered by IC. Kinetics studies with serum in the absence of cells suggested that interaction with RBC accelerated the rate of change in binding properties of IC. Rates of binding and release were independent of antigen/antibody ratio but were slowed and binding to RBC sustained when diluted or hypocomplementaemic (SLE) serum was substituted for neat serum. Our results suggest that competition for IC by RBC is associated with loss of ability of IC to bind to other blood cell types and reduction in size of IC, and that abnormalities of complement can lead to prolonged association of IC with RBC.


Assuntos
Complexo Antígeno-Anticorpo/imunologia , Receptores de Complemento/imunologia , Centrifugação com Gradiente de Concentração , Eritrócitos/imunologia , Humanos , Cinética , Leucócitos/imunologia , Neutrófilos/imunologia
14.
Proc Natl Acad Sci U S A ; 79(16): 5047-51, 1982 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-6214790

RESUMO

Antigen.antibody complexes (Ag.Ab) prepared from (125)I-labeled bovine serum albumin and guinea pig anti-albumin were incubated at 37 degrees C for 30 min with normal human serum diluted optimally for binding (1:16) and then with autologous erythrocytes (RBC). After washing, RBC-bearing antigen.antibody.complement complexes (Ag.Ab.C) were resuspended in serum reagents or solutions of purified complement components, and the kinetics of dissociation were analyzed. Ag.Ab.C dissociated in serum heated at 56 degrees C for 30 min (SDelta30) but not in serum heated for 120 min (SDelta120). Dissociation in SDelta30 markedly decreased after adsorption with anti-C3b inactivator but not anti-beta1H or anti-C4 binding protein (C4bp), and dissociation in SDelta120 markedly increased after addition of C3b inactivator. Hemolytic assays revealed that SDelta30 retained inactivator activity whereas SDelta120 lacked significant activity. Ag.Ab.C dissociated in the presence of purified inactivator or C3b but not beta1H or C3. Dissociation was more rapid with inactivator than with C3b and occurred at 0 degrees C as well as at 37 degrees C. Treatment with inactivator inhibitor abolished dissociation in SDelta30; dissociation in inactivator deficient serum was markedly reduced. Addition of beta1H did not enhance inactivator-mediated dissociation at limiting dilutions of inactivator, and adsorption of Ag.Ab.C with anti-beta1H or preparation of Ag.Ab.C with serum adsorbed with anti-beta1H did not diminish dissociation. After dissociation with inactivator, Ag.Ab.C were unchanged in size but were no longer able to bind to fresh RBC and gave enhanced binding to Raji and Daudi lymphoblastoid cells. NaDodSO(4)/polyacrylamide gel electrophoresis of Ag.Ab.C prepared with (125)I-labeled C3 revealed that, after binding to RBC, dissociation with inactivator was accompanied by generation of a C3 fragment the size of C3c. Preincubation of Ag.Ab.C with excess inactivator did not prevent subsequent binding of Ag.Ab.C to RBC but, immediately after binding, Ag.Ab.C dissociated rapidly. These findings indicate that C3b inactivator can release immune complexes from immune adherence receptors on human RBC, that release occurs independently of beta1H, alters cell binding properties of immune complexes, and involves multiple cleavages of the C3b alpha' chain, and that receptors in human RBC membrane are required for this C3b inactivator-mediated breakdown.


Assuntos
Complexo Antígeno-Anticorpo , Complemento C3/imunologia , Proteínas Inativadoras do Complemento C3b/imunologia , Membrana Eritrocítica/imunologia , Eritrócitos/imunologia , Receptores Imunológicos/metabolismo , Complemento C4/imunologia , Fator H do Complemento , Ligação Proteica , Solubilidade
15.
J Clin Lab Immunol ; 7(2): 93-103, 1982 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-7069786

RESUMO

Blood cells from patients with systemic lupus erythematosus (SLE) and from healthy control subjects were studied for their ability to bind exogenous immune complexes. Cells were mixed with experimental complexes (prepared from 125I-radiolabeled bovine serum albumin [BSA] and guinea pig antiserum to BSA) after incubation of the complexes with autologous serum in variable proportions. Unfractionated blood cells from SLE patients bound fewer added complexes (mean +/- SD = 39.8% +/- 12.9%) than cells from normal controls (mean +/- SD = 63.6% +/- 9.5%) (p less than 0.001); as much as 128-fold (mean = 23-fold) higher (p less than 0.01) proportions of serum to complex were required to optimize the lower binding. When SLE blood cells were mixed with complexes preincubated with normal sera instead of autologous SLE sera, SLE cells bound increased amounts of added complexes, and binding was optimal at the same lower proportions of serum to complex as for normal cells. When normal cells were mixed with complexes preincubated with compatible SLE sera in place of endogenous normal sera, binding to normal cells decreased to SLE levels, and the same higher proportions of serum to complex were required to optimize the lower binding as for the SLE cells. Similar results were obtained when binding to purified mononuclear cell preparations from SLE patients and controls was studied. When comparative studies were performed at high complex-to-cell ratios with complexes prepared using normal sera, SLE cells bound fewer complexes per cell than normal cells. These results indicate that although abnormalities in SLE cells are demonstrable, the major factor accounting for aberrant interaction between immune complexes and SLE cells resides in alterations in SLE sera.


Assuntos
Complexo Antígeno-Anticorpo/metabolismo , Lúpus Eritematoso Sistêmico/imunologia , Adolescente , Adulto , Células Sanguíneas/imunologia , Fenômenos Fisiológicos Sanguíneos , Proteínas do Sistema Complemento , Feminino , Humanos , Técnicas In Vitro , Lúpus Eritematoso Sistêmico/sangue , Masculino , Receptores Imunológicos
16.
J Immunol ; 130(3): 1336-40, 1983 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-6218206

RESUMO

We showed previously that soluble immune complexes (IC) added to normal human serum and unseparated autologous blood cells bind predominantly to red blood cells (RBC) bearing C3b receptors. RBC-associated IC are processed both by C3b/C4b inactivator (I) cleavage of C3b and by alternative pathway-dependent C3b generation, and are returned to the serum in a modified form. In the present study, soluble IC were prepared by using 125I-BSA and guinea pig anti-BSA, and were treated with diluted human serum to optimize sustained C3b-dependent binding to human RBC. This binding to RBC could not be abrogated by preincubation of IC with heat-inactivated human serum or purified serum regulatory proteins, I and beta 1H (H), but after binding, pretreated IC dissociated. When IC were prepared with serum containing 125I-C3, treatment with I + H did not release labeled C3 from the complexes. Analysis by polyacrylamide gel electrophoresis in sodium dodecyl sulfate (SDS-PAGE) of 125I-C3b in IC showed similar labeled peaks whether IC were pretreated with buffer or with I + H or were bound to RBC without pretreatment. In contrast, when IC containing 125I-C3b were exposed to I + RBC, nearly 70% of the label was released from the IC. The released material analyzed by SDS-PAGE showed major 125I-C3 peaks corresponding in m.w. to free beta-chain and alpha 1 cleavage products of 30,000 to 33,000 daltons. IC in undiluted serum after incubation with RBC gave less binding to human polymorphonuclear leukocytes than after incubation without RBC, but notably more binding to guinea pig spleen cells and human lymphoblastoid cells. These findings argue that exposure to serum regulatory proteins does not prevent subsequent binding of experimental IC to RBC, that RBC binding promotes degradation of C3b in IC which proceeds beyond iC3b, and that this I-mediated breakdown renders IC reactive with spleen cells.


Assuntos
Complexo Antígeno-Anticorpo/metabolismo , Proteínas Inativadoras do Complemento C3b/farmacologia , Eritrócitos/metabolismo , Baço/metabolismo , Animais , Sítios de Ligação , Bovinos , Proteínas Inativadoras do Complemento C3b/fisiologia , Fator H do Complemento , Cobaias , Humanos , Neutrófilos/metabolismo , Baço/citologia
17.
J Immunol ; 135(4): 2673-9, 1985 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-3161945

RESUMO

We have shown previously that C3b resides in a protected site when it is covalently bound to IgG (C3b-IgG). Such C3b displays a reduced affinity for factor H, with consequent enhanced survival in the presence of factors H and I and increased capacity for promoting alternative pathway consumption of C3. Because erythrocyte CR1 may be a major co-factor for factor I-mediated inactivation of immune complex-borne C3b in blood, we have examined the effect of covalently bound IgG on the C3b-CR1 interaction. Binding of monomeric C3b and C3b-IgG to human erythrocyte CR1 demonstrates identical ionic strength dependence for both species. Identical numbers of binding sites with indistinguishable affinities are detected by both ligands. Cleavage of the alpha'-chain of C3b and the alpha'-heavy chain of C3b-IgG proceeds at the same rate when erythrocyte CR1 serves as co-factor for factor I. Unlike factor H, CR1 supports a second cleavage of fluid-phase iC3b alpha'1 chain (free or bound to IgG) that generates C3c and a 33,000 m.w. fragment, which bears antigenic markers characteristic of C3g. Inactivation of C3b and C3b-IgG by CR1 and factor I also occurs at physiologic ionic strength, but proceeds very slowly relative to rates attainable with sub-physiologic inputs of factor H. CR1 does not recognize IgG-bound C3b as being in a protected site but, because of low binding affinity at physiologic ionic strength, is probably highly dependent on multivalent ligand-receptor interactions to efficiently exert its co-factor functions. Thus, inactivation of C3b-IgG heterodimers or small immune complexes bearing limited numbers of C3b residues may remain largely factor H-dependent in vivo, with resultant enhanced C3b survival.


Assuntos
Proteínas Inativadoras do Complemento C3b/metabolismo , Complemento C3b/metabolismo , Endopeptidases/fisiologia , Imunoglobulina G/fisiologia , Receptores de Complemento/metabolismo , Complexo Antígeno-Anticorpo/metabolismo , Sítios de Ligação , Proteínas Inativadoras do Complemento C3b/fisiologia , Fator H do Complemento , Fator I do Complemento , Endopeptidases/metabolismo , Eritrócitos/metabolismo , Humanos , Imunoglobulina G/metabolismo , Cinética , Concentração Osmolar , Receptores de Complemento 3b
18.
J Biol Chem ; 268(14): 9998-10002, 1993 May 15.
Artigo em Inglês | MEDLINE | ID: mdl-7683688

RESUMO

The glycoinositol phospholipid (GPI) anchors of mammalian proteins contain linear ethanolamine (EthN)-P-6ManManManGlcN glycan sequences that bear additional EthN-P substituents and in some cases include a fourth Man and a GalNAc or sialic acid-GalGalNAc. Precursors of these anchoring structures are preassembled in the endoplasmic reticulum by sequential glycosylation of inositol phospholipid. In previous studies (Hirose, S., Prince, G. M, Sevlever, D., Ravi, L., Rosenberry, T. L., Ueda, E., and Medof, M. E. (1992) J. Biol. Chem. 267, 16968-16974), a series of putative intermediates of this assembly process were isolated from human HeLa cells and murine lymphomas, and several of the more polar products were found to contain a second EthN-P attached to the Man residue (Man 1) proximal to GlcN. In this study, the most polar HeLa cell GPI species was purified by normal phase HeLa cell GPI species was purified by normal phase Iatrobead high performance liquid chromatography, and its glycan was characterized. Dionex anion exchange chromatographic analyses of fragments produced by nitrous acid deamination, hydrofluoric acid dephosphorylation, and trifluoroacetic acid hydrolysis in conjunction with biosynthetic labeling studies indicated a structure containing a third EthN-P substituent linked to the 6-position of Man 2. The polar GPI product exhibited a ManManManGlcN core lacking additional Man or GalNAc. The implications of the identification of this triply phosphoethanolamine-substituted species to mammalian GPI anchor biosynthesis are discussed.


Assuntos
Dolicol Monofosfato Manose/metabolismo , Glicosilfosfatidilinositóis/biossíntese , Microssomos/metabolismo , Fosfatidiletanolaminas/metabolismo , Sequência de Carboidratos , Cromatografia Líquida de Alta Pressão , Dolicol Monofosfato Manose/isolamento & purificação , Guanosina Difosfato Manose/metabolismo , Células HeLa , Humanos , Manose/metabolismo , Dados de Sequência Molecular , Polissacarídeos/isolamento & purificação , Polissacarídeos/metabolismo , Trítio , Uridina Difosfato N-Acetilglicosamina/metabolismo
19.
Clin Exp Immunol ; 87(3): 415-21, 1992 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-1371955

RESUMO

CD59, the membrane regulator of autologous C5b-9 channel formation, exhibits variable sensitivity to cleavage by phosphatidylinositol-specific phospholipase C (PI-PLC), an enzyme that releases glyco-inositolphospholipid (GPI)-anchored proteins from cell surfaces. To determine whether the GPI-anchor phospholipid of CD59 is similar to that of decay-accelerating factor (DAF) and whether variation in its structure underlies its variable enzyme susceptibility, the GPI anchors of the two proteins expressed on erythrocytes, polymorphonuclear and mononuclear leucocytes were compared in situ and after purification. Flow cytometric analyses of PI-PLC-treated cells showed parallel cell type specific release of both proteins as a function of enzyme concentration. Non-denaturing PAGE analyses of alkaline/hydroxylamine-treated proteins (affinity-purified from [125I]-surface-labelled cells) provided evidence for (i) comparable proportions of GPI-anchor acylation, and (ii) alkali-resistant rather than alkali-sensitive lipid substituents in erythrocytes. These findings argue that the differential C5b-9 sensitivity that distinguishes paroxysmal nocturnal haemoglobinuria II and III erythrocytes does not derive from expression of CD59 molecules with alternative GPI-anchor phospholipid structures.


Assuntos
Antígenos CD/química , Complexo de Ataque à Membrana do Sistema Complemento/antagonistas & inibidores , Glicolipídeos/química , Glicoproteínas de Membrana/química , Fosfatidilinositóis/química , Proteínas Sanguíneas/química , Antígenos CD55 , Antígenos CD59 , Proteínas Inativadoras do Complemento/química , Eletroforese em Gel de Poliacrilamida , Eritrócitos/efeitos dos fármacos , Citometria de Fluxo , Glicosilfosfatidilinositóis , Humanos , Leucócitos Mononucleares/efeitos dos fármacos , Proteínas de Membrana/química , Neutrófilos/efeitos dos fármacos , Relação Estrutura-Atividade , Fosfolipases Tipo C/farmacologia
20.
J Biol Chem ; 267(24): 16968-74, 1992 Aug 25.
Artigo em Inglês | MEDLINE | ID: mdl-1380957

RESUMO

A number of mammalian cell surface proteins are anchored by glycoinositol phospholipid (GPI) structures that are preassembled and transferred to them in the endoplasmic reticulum. The GPIs in these proteins contain linear ethanolamine (EthN)-phosphate (P)-6ManManManGlcN core glycan sequences bearing an additional EthN-P attached to the Man residue (Man 1) proximal to GlcN. The biochemical precursors of mammalian GPI anchor structures are incompletely characterized. In this study, putative [3H]Man-labeled GPI precursors were obtained by in vitro GDP-[3H] Man labeling of HeLa cell microsomes and by in vivo [3H]Man labeling of class B and F Thy-1 negative murine lymphoma mutants known to accumulate incomplete GPIs. The high performance liquid chromatography-purified in vitro and accumulated in vivo GPI products were structurally analyzed by nitrous acid deamination, hydrofluoric acid, trifluoroacetic acid hydrolysis, biosynthetic labeling, and exoglycosidase treatment. The data were consistent with a biosynthetic scheme in which Man and EthN-P are added stepwise to the developing glycan. Several additional points were demonstrated: 1) putative mammalian GPI precursors contain incomplete core glycans corresponding to those in previously characterized trypanosome GPI precursors. 2) The proximal EthN-P found in mature mammalian GPI anchor structures is added to Man 1 prior to incorporation of Man 2 and Man 3. 3) Glycans in the incomplete GPIs that accumulate in classes B and F lymphoma mutants consist of Man2- and Man3GlcN in which EthN-P is linked to Man 1. 4) Distal EthN-P linked to the 6-position of Man, characteristic of the complete GPI core, is found both in a subsequent GPI species with the glycan sequence EthN-P-6ManMan(EthN-P----)ManGlcN and in a more polar GPI product.


Assuntos
Etanolaminas/metabolismo , Glicolipídeos/metabolismo , Guanosina Difosfato Manose/metabolismo , Fosfatidilinositóis/metabolismo , Animais , Sequência de Carboidratos , Linhagem Celular , Cromatografia Líquida de Alta Pressão , Etanolamina , Glicosilfosfatidilinositóis , Células HeLa , Humanos , Linfoma , Proteínas de Membrana/metabolismo , Camundongos , Microssomos/metabolismo , Dados de Sequência Molecular
SELEÇÃO DE REFERÊNCIAS
Detalhe da pesquisa