Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 37
Filtrar
1.
J Hematol Oncol ; 15(1): 144, 2022 10 10.
Artículo en Inglés | MEDLINE | ID: mdl-36217194

RESUMEN

BACKGROUND: We assessed the mechanism by which multiple myeloma (MM) shapes the bone marrow (BM) microenvironment and affects MΦ polarization. METHODS: In vivo xenograft model of BM-disseminated human myeloma, as well as analysis of MM cell lines, stromal components, and primary samples from patients with MM, was utilized. RESULTS: Analysis of the BM from MM-bearing mice inoculated with human CXCR4-expressing RPMI8226 cells revealed a significant increase in M2 MΦ cell numbers (p < 0.01). CXCL13 was one of the most profoundly increased factors upon MM growth with increased levels in the blood of MM-bearing animals. Myeloid cells were the main source of the increased murine CXCL13 detected in MM-infiltrated BM. MM cell lines induced CXCL13 and concurrent expression of M2 markers (MERTK, CD206, CD163) in co-cultured human MΦ in vitro. Interaction with MΦ reciprocally induced CXCL13 expression in MM cell lines. Mechanistically, TGFß signaling was involved in CXCL13 induction in MM cells, while BTK signaling was implicated in MM-stimulated increase of CXCL13 in MΦ. Recombinant CXCL13 increased RANKL expression and induced TRAP+ osteoclast (OC) formation in vitro, while CXCL13 neutralization blocked these activities. Moreover, mice inoculated with CXCL13-silenced MM cells developed significantly lower BM disease. Reduced tumor load correlated with decreased numbers of M2 MΦ in BM, decreased bone disease, and lower expression of OC-associated genes. Finally, higher levels of CXCL13 were detected in the blood and BM samples of MM patients in comparison with healthy individuals. CONCLUSIONS: Altogether, our findings suggest that bidirectional interactions of MΦ with MM tumor cells result in M2 MΦ polarization, CXCL13 induction, and subsequent OC activation, enhancing their ability to support bone resorption and MM progression. CXCL13 may thus serve as a potential novel target in MM.


Asunto(s)
Quimiocina CXCL13 , Macrófagos , Mieloma Múltiple , Animales , Quimiocina CXCL13/metabolismo , Humanos , Macrófagos/metabolismo , Ratones , Mieloma Múltiple/patología , Factor de Crecimiento Transformador beta/metabolismo , Microambiente Tumoral , Tirosina Quinasa c-Mer/metabolismo
2.
J Hematol Oncol ; 13(1): 158, 2020 11 25.
Artículo en Inglés | MEDLINE | ID: mdl-33239060

RESUMEN

BACKGROUND: Chemoresistance remains a major treatment obstacle in multiple myeloma (MM). Novel new therapies are thus in need. Transient Receptor Potential Vanilloid type 1 (TRPV1) is a calcium-permeable ion channel that has been demonstrated to be expressed in solid tumors. Calcium channels have been shown to be involved in the regulation of cell proliferation, chemoresistance, migration and invasion. The aim of the current study was to evaluate its possible role in MM. METHODS: Pharmacological inhibitor was used to evaluate the role of TRPV1 in MM cell lines and primary MM cells. Flow cytometry, molecular analysis, fluorescent microscopy, proteomic analysis and xenograft in vivo model of MM with BM involvement were employed to assess the effect of TRPV1 inhibition and decipher its unique mechanism of action in MM. RESULTS: TRPV1 was found to be expressed by MM cell lines and primary MM cells. TRPV1 inhibition using the antagonist AMG9810-induced MM cell apoptosis and synergized with bortezomib, overcoming both CXCR4-dependent stroma-mediated and acquired resistance. In accordance, AMG9810 suppressed the expression and activation of CXCR4 in MM cells. TRPV1 inhibition increased mitochondrial calcium levels with subsequent mitochondrial ROS accumulation and depolarization. These effects were reversed by calcium chelation, suggesting the role of calcium perturbations in oxidative stress and mitochondrial destabilization. Furthermore, AMG9810 abolished bortezomib-induced accumulation of mitochondrial HSP70 and suppressed protective mitochondrial unfolded protein response. Proteomics revealed unique molecular signature related to the modification of ubiquitin signaling pathway. Consequently, 38 proteins related to the ubiquitylation machinery were downregulated upon combined bortezomib/AMG9810 treatment. Concomitantly, AMG9810 abolished bortezomib-induced ubiquitination of cytosolic and mitochondrial proteins. Furthermore, bortezomib/AMG9810 treatment induced mitochondrial accumulation of PINK1, significantly reduced the mitochondrial mass and promoted mitochondrial-lysosomal fusion, indicating massive mitophagy. Finally, in a recently developed xenograft model of systemic MM with BM involvement, bortezomib/AMG9810 treatment effectively reduced tumor burden in the BM of MM-bearing mice. CONCLUSIONS: Altogether, our results unravel the mechanism mediating the strong synergistic anti-MM activity of bortezomib in combination with TRPV1 inhibition which may be translated into the clinic.


Asunto(s)
Acrilamidas/farmacología , Antineoplásicos/farmacología , Bortezomib/farmacología , Compuestos Bicíclicos Heterocíclicos con Puentes/farmacología , Mitofagia/efectos de los fármacos , Mieloma Múltiple/tratamiento farmacológico , Canales Catiónicos TRPV/antagonistas & inhibidores , Acrilamidas/uso terapéutico , Animales , Antineoplásicos/uso terapéutico , Bortezomib/uso terapéutico , Compuestos Bicíclicos Heterocíclicos con Puentes/uso terapéutico , Calcio/metabolismo , Línea Celular Tumoral , Humanos , Ratones , Mieloma Múltiple/metabolismo , Canales Catiónicos TRPV/metabolismo , Ubiquitina/metabolismo , Respuesta de Proteína Desplegada/efectos de los fármacos
3.
Sci Rep ; 9(1): 12876, 2019 10 02.
Artículo en Inglés | MEDLINE | ID: mdl-31578341

RESUMEN

An autoimmune response against myelin protein is considered one of the key pathogenic processes that initiates multiple sclerosis (MS). The currently available MS disease modifying therapies have demonstrated to reduce the frequency of inflammatory attacks. However, they appear limited in preventing disease progression and neurodegeneration. Hence, novel therapeutic approaches targeting both inflammation and neuroregeneration are urgently needed. A new pregnancy derived synthetic peptide, synthetic PreImplantation Factor (sPIF), crosses the blood-brain barrier and prevents neuro-inflammation. We report that sPIF reduces paralysis and de-myelination of the brain in a clinically-relevant experimental autoimmune encephalomyelitis mice model. These effects, at least in part, are due to post-translational modifications, which involve cyclic AMP dependent protein kinase (PKA), calcium-dependent protein kinase (PKC), and immune regulation. In terms of potential MS treatment, sPIF was successfully tested in neurodegenerative animal models of perinatal brain injury and experimental autoimmune encephalitis. Importantly, sPIF received a FDA Fast Track Approval for first in human trial in autommuninty (completed).


Asunto(s)
Encefalomielitis Autoinmune Experimental , Esclerosis Múltiple , Parálisis , Péptidos , Procesamiento Proteico-Postraduccional/efectos de los fármacos , Animales , Encefalomielitis Autoinmune Experimental/tratamiento farmacológico , Encefalomielitis Autoinmune Experimental/metabolismo , Encefalomielitis Autoinmune Experimental/patología , Femenino , Ratones , Esclerosis Múltiple/tratamiento farmacológico , Esclerosis Múltiple/metabolismo , Esclerosis Múltiple/patología , Parálisis/tratamiento farmacológico , Parálisis/metabolismo , Parálisis/patología , Péptidos/farmacocinética , Péptidos/farmacología
4.
Biochem Pharmacol ; 168: 412-428, 2019 10.
Artículo en Inglés | MEDLINE | ID: mdl-31325448

RESUMEN

Although having promising anti-myeloma properties, the pan-histone deacetylase inhibitor (HDACi) panobinostat lacks therapeutic activity as a single agent. The aim of the current study was to elucidate the mechanisms underlying multiple myeloma (MM) resistance to panobinostat monotherapy and to define strategies to overcome it. Sensitivity of MM cell lines and primary CD138+ cells from MM patients to panobinostat correlated with reduced expression of the chemokine receptor CXCR4, whereas overexpression of CXCR4 in MM cell lines increased their resistance to panobinostat. Decreased sensitivity to HDACi was associated with reversible G0/G1 cell growth arrest while response was characterized by apoptotic cell death. Analysis of intra-cellular signaling mediators revealed the pro-survival mTOR pathway to be regulated by CXCR4 overexpression. Combining panobinostat with mTOR inhibitor everolimus abrogated the resistance to HDACi and induced synergistic cell death. The combination of panobinostat/everolimus resulted in sustained DNA damage and irreversible suppression of proliferation accompanied by robust apoptosis. Gene expression analysis revealed distinct genetic profiles of single versus combined agent exposure. Whereas panobinostat increased the expression of the cell cycle inhibitor p21, co-treatment with everolimus abrogated the increase in p21 and synergistically downregulated the expression of DNA repair genes and mitotic checkpoint regulators. Importantly, the combination of panobinostat with everolimus effectively targeted CXCR4-expressing resistant MM cells in vivo in the BM niche. In summary, our results uncover the mechanism responsible for the strong synergistic anti-MM activity of dual HDAC and mTOR inhibition and provide the rationale for a novel potential therapeutic approach to treat MM.


Asunto(s)
Antineoplásicos/administración & dosificación , Everolimus/administración & dosificación , Inhibidores de Histona Desacetilasas/administración & dosificación , Mitosis/efectos de los fármacos , Panobinostat/administración & dosificación , Receptores CXCR4/antagonistas & inhibidores , Proteínas de Unión al GTP rho/antagonistas & inhibidores , Animales , Células de la Médula Ósea/efectos de los fármacos , Células de la Médula Ósea/metabolismo , Línea Celular Tumoral , Células Cultivadas , Relación Dosis-Respuesta a Droga , Resistencia a Antineoplásicos/efectos de los fármacos , Resistencia a Antineoplásicos/fisiología , Humanos , Ratones , Mitosis/fisiología , Mieloma Múltiple/tratamiento farmacológico , Mieloma Múltiple/metabolismo , Receptores CXCR4/biosíntesis , Receptores CXCR4/genética , Serina-Treonina Quinasas TOR/antagonistas & inhibidores , Serina-Treonina Quinasas TOR/biosíntesis , Serina-Treonina Quinasas TOR/genética , Ensayos Antitumor por Modelo de Xenoinjerto/métodos , Proteínas de Unión al GTP rho/biosíntesis , Proteínas de Unión al GTP rho/genética
5.
Cancer Res ; 78(6): 1471-1483, 2018 03 15.
Artículo en Inglés | MEDLINE | ID: mdl-29259008

RESUMEN

CXCR4 expression in neuroblastoma tumors correlates with disease severity. In this study, we describe mechanisms by which CXCR4 signaling controls neuroblastoma tumor growth and response to therapy. We found that overexpression of CXCR4 or stimulation with CXCL12 supports neuroblastoma tumorigenesis. Moreover, CXCR4 inhibition with the high-affinity CXCR4 antagonist BL-8040 prevented tumor growth and reduced survival of tumor cells. These effects were mediated by the upregulation of miR-15a/16-1, which resulted in downregulation of their target genes BCL-2 and cyclin D1, as well as inhibition of ERK. Overexpression of miR-15a/16-1 in cells increased cell death, whereas antagomirs to miR-15a/16-1 abolished the proapoptotic effects of BL-8040. CXCR4 overexpression also increased miR-15a/16-1, shifting their oncogenic dependency from the BCL-2 to the ERK signaling pathway. Overall, our results demonstrate the therapeutic potential of CXCR4 inhibition in neuroblastoma treatment and provide a rationale to test combination therapies employing CXCR4 and BCL-2 inhibitors to increase the efficacy of these agents.Significance: These results provide a mechanistic rationale for combination therapy of CXCR4 and BCL-2 inhibitors to treat a common and commonly aggressive pediatric cancer.Cancer Res; 78(6); 1471-83. ©2017 AACR.


Asunto(s)
Neoplasias Encefálicas/patología , MicroARNs/metabolismo , Neuroblastoma/patología , Receptores CXCR4/metabolismo , Animales , Neoplasias Encefálicas/tratamiento farmacológico , Neoplasias Encefálicas/genética , Línea Celular Tumoral , Ciclina D1/genética , Ciclina D1/metabolismo , Resistencia a Antineoplásicos/efectos de los fármacos , Resistencia a Antineoplásicos/genética , Regulación Neoplásica de la Expresión Génica , Humanos , Sistema de Señalización de MAP Quinasas/efectos de los fármacos , Sistema de Señalización de MAP Quinasas/genética , Ratones , MicroARNs/genética , Neuroblastoma/tratamiento farmacológico , Neuroblastoma/genética , Péptidos/farmacología , Proteínas Proto-Oncogénicas c-bcl-2/genética , Proteínas Proto-Oncogénicas c-bcl-2/metabolismo , Receptores CXCR4/antagonistas & inhibidores , Receptores CXCR4/genética , Ensayos Antitumor por Modelo de Xenoinjerto
6.
Front Immunol ; 8: 1432, 2017.
Artículo en Inglés | MEDLINE | ID: mdl-29218043

RESUMEN

Chemokines and their receptors play critical roles in the progression of autoimmunity and inflammation. Typically, multiple chemokines are involved in the development of these pathologies. Indeed, targeting single chemokines or chemokine receptors has failed to achieve significant clinical benefits in treating autoimmunity and inflammation. Moreover, the binding of host atypical chemokine receptors to multiple chemokines as well as the binding of chemokine-binding proteins secreted by various pathogens can serve as a strategy for controlling inflammation. In this work, promiscuous chemokine-binding peptides that could bind and inhibit multiple inflammatory chemokines, such as CCL2, CCL5, and CXCL9/10/11, were selected from phage display libraries. These peptides were cloned into human mutated immunoglobulin Fc-protein fusions (peptibodies). The peptibodies BKT120Fc and BKT130Fc inhibited the ability of inflammatory chemokines to induce the adhesion and migration of immune cells. Furthermore, BKT120Fc and BKT130Fc also showed a significant inhibition of disease progression in a variety of animal models for autoimmunity and inflammation. Developing a novel class of antagonists that can control the courses of diseases by selectively blocking multiple chemokines could be a novel way of generating effective therapeutics.

7.
Clin Cancer Res ; 23(7): 1733-1747, 2017 Apr 01.
Artículo en Inglés | MEDLINE | ID: mdl-27697999

RESUMEN

Purpose: To explore the functional consequences of possible cross-talk between the CXCR4/CXCL12 and the sphingosine-1-phosphate (S1P) pathways in multiple myeloma (MM) cells and to evaluate the effect of S1P targeting with the FTY720 modulator as a potential anti-MM therapeutic strategy.Experimental Design and Results: S1P targeting with FTY720 induces MM cell apoptosis. The combination of FTY720 with the SPHK1 inhibitor SKI-II results in synergistic inhibition of MM growth. CXCR4/CXCL12-enhanced expression correlates with reduced MM cell sensitivity to both FTY720 and SKI-II inhibitors, and with SPHK1 coexpression in both cell lines and primary MM bone marrow (BM) samples, suggesting regulative cross-talk between the CXCR4/CXCL12 and SPHK1 pathways in MM cells. FTY720 was found to directly target CXCR4. FTY720 profoundly reduces CXCR4 cell-surface levels and abrogates the CXCR4-mediated functions of migration toward CXCL12 and signaling pathway activation. Moreover, FTY720 cooperates with bortezomib, inducing its cytotoxic activity and abrogating the bortezomib-mediated increase in CXCR4 expression. FTY720 effectively targets bortezomib-resistant cells and increases their sensitivity to bortezomib, promoting DNA damage. Finally, in a recently developed novel xenograft model of CXCR4-dependent systemic MM with BM involvement, FTY720 treatment effectively reduces tumor burden in the BM of MM-bearing mice. FTY720 in combination with bortezomib demonstrates superior tumor growth inhibition and abrogates bortezomib-induced CXCR4 increase on MM cells.Conclusions: Altogether, our work identifies a cross-talk between the S1P and CXCR4 pathways in MM cells and provides a preclinical rationale for the therapeutic application of FTY720 in combination with bortezomib in patients with MM. Clin Cancer Res; 23(7); 1733-47. ©2016 AACR.


Asunto(s)
Quimiocina CXCL12/genética , Mieloma Múltiple/tratamiento farmacológico , Fosfotransferasas (Aceptor de Grupo Alcohol)/genética , Receptores CXCR4/genética , Animales , Apoptosis/efectos de los fármacos , Bortezomib/administración & dosificación , Daño del ADN/efectos de los fármacos , Sinergismo Farmacológico , Clorhidrato de Fingolimod/administración & dosificación , Regulación Neoplásica de la Expresión Génica/efectos de los fármacos , Humanos , Lisofosfolípidos/genética , Lisofosfolípidos/metabolismo , Ratones , Mieloma Múltiple/genética , Mieloma Múltiple/patología , Fosfotransferasas (Aceptor de Grupo Alcohol)/antagonistas & inhibidores , Transducción de Señal/efectos de los fármacos , Esfingosina/análogos & derivados , Esfingosina/genética , Esfingosina/metabolismo , Ensayos Antitumor por Modelo de Xenoinjerto
8.
PLoS One ; 10(12): e0143589, 2015.
Artículo en Inglés | MEDLINE | ID: mdl-26624007

RESUMEN

CD44 is a multi-functional receptor with multiple of isoforms engaged in modulation of cell trafficking and transmission of apoptotic signals. We have previously shown that injection of anti-CD44 antibody into NOD mice induced resistance to type 1 diabetes (T1D). In this communication we describe our efforts to understand the mechanism underlying this effect. We found that CD44-deficient NOD mice develop stronger resistance to T1D than wild-type littermates. This effect is not explained by the involvement of CD44 in cell migration, because CD44-deficient inflammatory cells surprisingly had greater invasive potential than the corresponding wild type cells, probably owing to molecular redundancy. We have previously reported and we show here again that CD44 expression and hyaluronic acid (HA, the principal ligand for CD44) accumulation are detected in pancreatic islets of diabetic NOD mice, but not of non-diabetic DBA/1 mice. Expression of CD44 on insulin-secreting ß cells renders them susceptible to the autoimmune attack, and is associated with a diminution in ß-cells function (e.g., less insulin production and/or insulin secretion) and possibly also with an enhanced apoptosis rate. The diabetes-supportive effect of CD44 expression on ß cells was assessed by the TUNEL assay and further strengthened by functional assays exhibiting increased nitric oxide release, reduced insulin secretion after glucose stimulation and decreased insulin content in ß cells. All these parameters could not be detected in CD44-deficient islets. We further suggest that HA-binding to CD44-expressing ß cells is implicated in ß-cell demise. Altogether, these data agree with the concept that CD44 is a receptor capable of modulating cell fate. This finding is important for other pathologies (e.g., cancer, neurodegenerative diseases) in which CD44 and HA appear to be implicated.


Asunto(s)
Diabetes Mellitus Tipo 1/metabolismo , Diabetes Mellitus Tipo 1/patología , Receptores de Hialuranos/metabolismo , Células Secretoras de Insulina/patología , Animales , Apoptosis , Transporte Biológico , Movimiento Celular , Diabetes Mellitus Tipo 1/genética , Femenino , Regulación de la Expresión Génica , Glucosa/metabolismo , Receptores de Hialuranos/genética , Ácido Hialurónico/metabolismo , Ratones
9.
PLoS One ; 8(8): e72722, 2013.
Artículo en Inglés | MEDLINE | ID: mdl-23991146

RESUMEN

Cerebral malaria is the most severe complication of Plasmodium falciparum infection, and a leading cause of death in children under the age of five in malaria-endemic areas. We report high therapeutic efficacy of a novel formulation of liposome-encapsulated water-soluble glucocorticoid prodrugs, and in particular ß-methasone hemisuccinate (BMS), for treatment of experimental cerebral malaria (ECM), using the murine P. berghei ANKA model. BMS is a novel derivative of the potent steroid ß-methasone, and was specially synthesized to enable remote loading into nano-sterically stabilized liposomes (nSSL), to form nSSL-BMS. The novel nano-drug, composed of nSSL remote loaded with BMS, dramatically improves drug efficacy and abolishes the high toxicity seen upon administration of free BMS. nSSL-BMS reduces ECM rates in a dose-dependent manner and creates a survival time-window, enabling administration of an antiplasmodial drug, such as artemisone. Administration of artemisone after treatment with the nSSL-BMS results in complete cure. Treatment with BMS leads to lower levels of cerebral inflammation, demonstrated by changes in cytokines, chemokines, and cell markers, as well as diminished hemorrhage and edema, correlating with reduced clinical score. Administration of the liposomal formulation results in accumulation of BMS in the brains of sick mice but not of healthy mice. This steroidal nano-drug effectively eliminates the adverse effects of the cerebral syndrome even when the treatment is started at late stages of disease, in which disruption of the blood-brain barrier has occurred and mice show clear signs of neurological impairment. Overall, sequential treatment with nSSL-BMS and artemisone may be an efficacious and well-tolerated therapy for prevention of CM, elimination of parasites, and prevention of long-term cognitive damage.


Asunto(s)
Betametasona/uso terapéutico , Modelos Animales de Enfermedad , Liposomas , Malaria Cerebral/tratamiento farmacológico , Nanopartículas , Enfermedad Aguda , Animales , Secuencia de Bases , Betametasona/administración & dosificación , Cartilla de ADN , Malaria Cerebral/parasitología , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Endogámicos ICR , Plasmodium berghei/aislamiento & purificación , Reacción en Cadena en Tiempo Real de la Polimerasa
10.
Clin Cancer Res ; 19(13): 3495-507, 2013 Jul 01.
Artículo en Inglés | MEDLINE | ID: mdl-23637121

RESUMEN

PURPOSE: Chemokine axis CXCR4/CXCL12 is critically involved in the survival and trafficking of normal and malignant B lymphocytes. Here, we investigated the effect of high-affinity CXCR4 antagonist BKT140 on lymphoma cell growth and rituximab-induced cytotoxicity in vitro and in vivo. EXPERIMENTAL DESIGN: In vitro efficacy of BKT140 alone or in combination with rituximab was determined in non-Hodgkin lymphoma (NHL) cell lines and primary samples from bone marrow aspirates of patients with NHL. In vivo efficacy was evaluated in xenograft models of localized and disseminated NHL with bone marrow involvement. RESULTS: Antagonizing CXCR4 with BKT140 resulted in significant inhibition of CD20+ lymphoma cell growth and in the induction of cell death, respectively. Combination of BKT140 with rituximab significantly enhanced the apoptosis against the lymphoma cells in a dose-dependent manner. Moreover, rituximab induced CXCR4 expression in lymphoma cell lines and primary lymphoma cells, suggesting the possible interaction between CD20 and CXCR4 pathways in NHL. Primary bone marrow stromal cells (BMSC) further increased CXCR4 expression and protected NHL cells from rituximab-induced apoptosis, whereas BKT140 abrogated this protective effect. Furthermore, BKT140 showed efficient antilymphoma activity in vivo in the xenograft model of disseminated NHL with bone marrow involvement. BKT140 treatment inhibited the local tumor progression and significantly reduced the number of NHL cells in the bone marrow. Combined treatment of BKT140 with rituximab further decreased the number of viable lymphoma cells in the bone marrow, achieving 93% reduction. CONCLUSIONS: These findings suggest the possible role of CXCR4 in NHL progression and response to rituximab and provide the scientific basis for the development of novel CXCR4-targeted therapies for refractory NHL.


Asunto(s)
Anticuerpos Monoclonales de Origen Murino/farmacología , Antígenos CD20/metabolismo , Antineoplásicos/farmacología , Linfoma no Hodgkin/metabolismo , Oligopéptidos/farmacología , Receptores CXCR4/metabolismo , Transducción de Señal/efectos de los fármacos , Animales , Apoptosis/efectos de los fármacos , Médula Ósea/patología , Ciclo Celular/efectos de los fármacos , Línea Celular Tumoral , Proliferación Celular/efectos de los fármacos , Microambiente Celular/efectos de los fármacos , Humanos , Linfoma no Hodgkin/tratamiento farmacológico , Linfoma no Hodgkin/patología , Ratones , Rituximab , Ensayos Antitumor por Modelo de Xenoinjerto
11.
Am J Reprod Immunol ; 68(6): 456-64, 2012 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-22805418

RESUMEN

A typical autoimmune neuro-inflammatory disease (NID), multiple sclerosis (MS), is more prevalent in women than in men. Majority of patients with MS are of child-bearing age; therefore, occurrence in pregnancy is common. Herein, we review proposed disease mechanisms and suggest therapeutic interventions, focusing on the remarkable pregnancy-induced protection against MS - insofar considered as best, albeit temporary therapy for such harsh NID. Current drugs used for MS therapy in pregnancy are described. Role of non-pregnancy-specific agents considered involved in amelioration of disease is also presented. This review highlights pregnancy-derived neuro-protective agents, proposing that unique pregnancy-induced immune-protective environment is because of the conceptus and its direct action. The essential role of pre-implantation factor (PIF) in pregnancy is delineated. Finally, PIF immune-modulatory effects and efficacy in chronic model of neuro-inflammation to reduce inflammation and paralysis coupled with neural regeneration is presented. Overall, we postulate that this embryo-derived-compound holds great promise to improve MS and possibly neuro-inflammation in general.


Asunto(s)
Esclerosis Múltiple/tratamiento farmacológico , Esclerosis Múltiple/inmunología , Péptidos/uso terapéutico , Femenino , Humanos , Inflamación , Masculino , Regeneración Nerviosa , Fármacos Neuroprotectores/metabolismo , Parálisis , Embarazo , Complicaciones del Embarazo/tratamiento farmacológico , Células TH1/metabolismo , Células Th2/metabolismo , Deficiencia de Vitamina D , alfa-Fetoproteínas/uso terapéutico
12.
J Neurol Sci ; 312(1-2): 146-57, 2012 Jan 15.
Artículo en Inglés | MEDLINE | ID: mdl-21996270

RESUMEN

INTRODUCTION: Embryo-derived PIF modulates systemic maternal immunity without suppression. Synthetic analog (sPIF) prevents juvenile diabetes, preserves islet function, reducing oxidative stress/protein misfolding. We investigate sPIF effectiveness in controlling neuroinflammation/MS. METHODS: Examine sPIF-induced protection against harsh, clinical-relevant murine EAE-PLP acute and chronic models. Evaluate clinical indices: circulating cytokines, spinal cord histology, genome, canonical global proteome, cultured PLP-activated splenocytes cytokines, and immunophenotype. RESULTS: Short-term, low-dose sPIF prevented paralysis development and lowered mortality (P<0.05). Episodic sPIF reversed chronic paralysis (P<0.0001) completely in >50%, by day 82. Prevention model: 12days post-therapy, sPIF reduced circulating IL12 ten-fold and inflammatory cells access to spinal cord. Regression model: sPIF blocked PLP-induced IL17 and IL6 secretions. Long-term chronic model: sPIF reduced spinal cord pro-inflammatory cytokines/chemokines, (ALCAM, CF1, CCL8), apoptosis-promoters, inflammatory cells access (JAM3, OPA1), solute channels (ATPases), aberrant coagulation factors (Serpins), and pro-antigenic MOG. Canonical proteomic analysis demonstrated reduced oxidative phosphorylation, vesicle traffic, cytoskeleton remodeling involved in neuro-cytoskeleton breakdown (tubulins), associated with axon re-assembly by (MTAPs)/improved synaptic transmission. CONCLUSION: sPIF--through coordinated central and systemic multi-targeted action--reverses neuroinflammation/MS and imparts significant neuroprotective effects up to total paralysis resolution. Clinical testing is warranted and planned.


Asunto(s)
Encefalomielitis Autoinmune Experimental/tratamiento farmacológico , Encefalomielitis Autoinmune Experimental/patología , Mediadores de Inflamación/farmacología , Regeneración Nerviosa/efectos de los fármacos , Péptidos/farmacología , Animales , Modelos Animales de Enfermedad , Encefalomielitis Autoinmune Experimental/inmunología , Femenino , Mediadores de Inflamación/uso terapéutico , Ratones , Ratones Endogámicos , Esclerosis Múltiple/tratamiento farmacológico , Esclerosis Múltiple/inmunología , Esclerosis Múltiple/patología , Regeneración Nerviosa/inmunología , Péptidos/uso terapéutico , Distribución Aleatoria
13.
Endocrine ; 40(1): 41-54, 2011 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-21424847

RESUMEN

Preimplantation factor (PIF) is a novel embryo-secreted immunomodulatory peptide. Its synthetic analog (sPIF) modulates maternal immunity without suppression. There is an urgent need to develop agents that could prevent the development of type 1 diabetes mellitus (TIDM). Herein, we examine sPIF's preventive effect on TIDM development by using acute adoptive-transfer (ATDM) and spontaneously developing (SDM) in non-obese diabetic (NOD) murine models. Diabetes was evaluated by urinary and plasma glucose, intraperitoneal glucose tolerance test (IPGTT), pancreatic islets insulin staining by immunohistochemistry and by pancreatic proteome evaluation using mass spectrometry, followed by signal pathway analysis. Continuous administration of sPIF for 4-weeks prevents diabetes development in ATDM model in >90% of recipients demonstrated by normal IPGTT, preserved islets architecture, number, and insulin staining. (P < 0.01). sPIF effect was specific; its protective effects are not replicated by scrambled PIF (χ(2) = 0.009) control. sPIF led also to increased circulating Th2 and Th1 cytokines. In SDM model, 4-week continuous sPIF administration prevented onset of diabetes for 21 weeks post-therapy (P < 0.01). Low-dose sPIF administration for 16 weeks prevented diabetes development up to 14 weeks post-therapy, evidenced by preserved islets architecture and insulin staining. In SDM model, pancreatic proteome pathway analysis demonstrated that sPIF regulates protein traffic, prevents protein misfolding and aggregation, and reduces oxidative stress and islets apoptosis, leading to preserved insulin staining. sPIF further increased insulin receptor expression and reduced actin and tubulin proteins, thereby blocking neutrophil invasion and inflammation. Exocrine pancreatic function was also preserved. sPIF administration results in marked prevention of spontaneous and induced adoptive-transfer diabetes suggesting its potential effectiveness in treating early-stage TIDM.


Asunto(s)
Diabetes Mellitus Tipo 1/prevención & control , Páncreas/efectos de los fármacos , Páncreas/fisiología , Péptidos/farmacología , Péptidos/uso terapéutico , Animales , Apoptosis/efectos de los fármacos , Citocinas/sangre , Diabetes Mellitus Tipo 1/metabolismo , Modelos Animales de Enfermedad , Relación Dosis-Respuesta a Droga , Implantes de Medicamentos , Femenino , Glucosa/metabolismo , Insulina/metabolismo , Islotes Pancreáticos/metabolismo , Masculino , Ratones , Ratones Endogámicos NOD , Estrés Oxidativo/efectos de los fármacos , Páncreas/metabolismo , Péptidos/administración & dosificación
14.
Mol Immunol ; 47(10): 1890-8, 2010 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-20399501

RESUMEN

Heparanase is an endo-beta-d-glucuronidase that cleaves heparan sulfate (HS) saccharide chains. The enzyme promotes cell adhesion, migration and invasion and plays a significant role in cancer metastasis, angiogenesis and inflammation. The present study focuses on the involvement of heparanase in autoimmunity, applying the murine experimental autoimmune encephalitis (EAE) model, a T-cell dependent disease often used to investigate the pathophysiology of multiple sclerosis (MS). Intraperitoneal administration of recombinant heparanase ameliorated, in a dose dependent manner, the clinical signs of the disease. In vitro and in vivo studies revealed that heparanase inhibited mitogen induced splenocyte proliferation and mixed lymphocyte reaction (MLR) through modulation of their repertoire of cytokines indicated by a marked increase in the levels of IL-4, IL-6 and IL-10, and a parallel decrease in IL-12 and TNF-alpha. Similar results were obtained with active, latent, or point mutated inactive heparanase, indicating that the observed inhibitory effect is attributed to a non-enzymatic activity of the heparanase protein. We suggest that heparanase induces upregulation of Th2 cytokines, resulting in inhibition of the inflammatory lesion of EAE.


Asunto(s)
Citocinas/inmunología , Encefalomielitis Autoinmune Experimental/inmunología , Glucuronidasa/metabolismo , Activación de Linfocitos/inmunología , Células Th2/inmunología , Animales , Citocinas/biosíntesis , Encefalomielitis Autoinmune Experimental/enzimología , Encefalomielitis Autoinmune Experimental/patología , Ensayo de Inmunoadsorción Enzimática , Femenino , Glucuronidasa/inmunología , Prueba de Cultivo Mixto de Linfocitos , Ratones , Médula Espinal/patología , Células Th2/metabolismo , Regulación hacia Arriba
15.
PLoS One ; 5(4): e10135, 2010 Apr 15.
Artículo en Inglés | MEDLINE | ID: mdl-20419162

RESUMEN

BACKGROUND: Heparanase, endoglycosidase that cleaves heparan sulfate side chains of heparan sulfate proteoglycans, plays important roles in cancer metastasis, angiogenesis and inflammation. DESIGN AND METHODS: Applying a mouse model of bone marrow transplantation and transgenic mice over-expressing heparanase, we evaluated the effect of heparanase on the engraftment process and the development of graft-versus-host disease. RESULTS: Analysis of F1 mice undergoing allogeneic bone marrow transplantation from C57BL/6 mice demonstrated a better and faster engraftment in mice receiving cells from donors that were pretreated with heparanase. Moreover, heparanase treated recipient F1 mice showed only a mild appearance of graft-versus-host disease and died 27 days post transplantation while control mice rapidly developed signs of graft-versus-host disease (i.e., weight loss, hair loss, diarrhea) and died after 12 days, indicating a protective effect of heparanase against graft-versus-host disease. Similarly, we applied transgenic mice over-expressing heparanase in most tissues as the recipients of BMT from C57BL/6 mice. Monitoring clinical parameters of graft-versus-host disease, the transgenic mice showed 100% survival on day 40 post transplantation, compared to only 50% survival on day 14, in the control group. In vitro and in vivo studies revealed that heparanase inhibited T cell function and activation through modulation of their cytokine repertoire, indicated by a marked increase in the levels of Interleukin-4, Interleukin-6 and Interleukin-10, and a parallel decrease in Interleukin-12, tumor necrosis factor-alfa and interferon-gamma. Using point mutated inactive enzyme, we found that the shift in cytokine profile was independent of heparanase enzymatic activity. CONCLUSIONS: Our results indicate a significant role of heparanase in bone marrow transplantation biology, facilitating engraftment and suppressing graft-versus-host disease, apparently through an effect on T cell activation and cytokine production pattern.


Asunto(s)
Trasplante de Médula Ósea/métodos , Glucuronidasa/farmacología , Supervivencia de Injerto/efectos de los fármacos , Enfermedad Injerto contra Huésped/prevención & control , Animales , Trasplante de Médula Ósea/efectos adversos , Citocinas/biosíntesis , Citocinas/efectos de los fármacos , Glucuronidasa/administración & dosificación , Glucuronidasa/uso terapéutico , Ratones , Ratones Endogámicos , Ratones Transgénicos , Tasa de Supervivencia , Linfocitos T/efectos de los fármacos , Linfocitos T/inmunología
16.
PLoS One ; 5(2): e9132, 2010 Feb 09.
Artículo en Inglés | MEDLINE | ID: mdl-20161752

RESUMEN

BACKGROUND: Telomere/telomerase system has been recently recognized as an attractive target for anticancer therapy. Telomerase inhibition results in tumor regression and increased sensitivity to various cytotoxic drugs. However, it has not been fully established yet whether the mediator of these effects is telomerase inhibition per se or telomere shortening resulting from inhibition of telomerase activity. In addition, the characteristics and mechanisms of sensitization to cytotoxic drugs caused by telomerase inhibition has not been elucidated in a systematic manner. METHODOLOGY/PRINCIPAL FINDINGS: In this study we characterized the relative importance of telomerase inhibition versus telomere shortening in cancer cells. Sensitization of cancer cells to cytotoxic drugs was achieved by telomere shortening in a length dependent manner and not by telomerase inhibition per se. In our system this sensitization was related to the mechanism of action of the cytotoxic drug. In addition, telomere shortening affected also other cancer cell functions such as migration. Telomere shortening induced DNA damage whose repair was impaired after administration of cisplatinum while doxorubicin or vincristine did not affect the DNA repair. These findings were verified also in in vivo mouse model. The putative explanation underlying the phenotype induced by telomere shortening may be related to changes in expression of various microRNAs triggered by telomere shortening. CONCLUSIONS/SIGNIFICANCE: To our best knowledge this is the first study characterizing the relative impact of telomerase inhibition and telomere shortening on several aspects of cancer cell phenotype, especially related to sensitivity to cytotoxic drugs and its putative mechanisms. The microRNA changes in cancer cells upon telomere shortening are novel information. These findings may facilitate the development of telomere based approaches in treatment of cancer.


Asunto(s)
Antineoplásicos/farmacología , Neoplasias/tratamiento farmacológico , Neoplasias/genética , Telómero/genética , Animales , Apoptosis/efectos de los fármacos , Línea Celular Tumoral , Movimiento Celular/efectos de los fármacos , Supervivencia Celular/efectos de los fármacos , Cisplatino/farmacología , Ensayo Cometa , Daño del ADN , Reparación del ADN , Relación Dosis-Respuesta a Droga , Femenino , Humanos , Células K562 , Masculino , Ratones , Ratones Endogámicos BALB C , Ratones Desnudos , Neoplasias/patología , Oligonucleótidos/farmacología , Telomerasa/antagonistas & inhibidores , Telomerasa/genética , Telomerasa/metabolismo , Telómero/metabolismo , Ensayos Antitumor por Modelo de Xenoinjerto
17.
Cancer Invest ; 28(5): 535-43, 2010 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-20014940

RESUMEN

The anticancer effects of synthetic, non-natural analogs of ceramide were tested using human TSU-Pr1 prostate cancer cells in-vitro as well as in-vivo, following their effects on tumors development in mice. When incubated with the cultured cancer cells, the analogs elevated cellular ceramide and induced a cytotoxicity and death by apoptosis. When a ceramide analog was injected intradermally or intraperitoneally into BALB/c-Nude or NOD-SCID mice bearing a human prostate tumor, a considerable regression of the tumor was observed. The synthetic ceramide analogs should thus be further investigated as potential anticancer drugs.


Asunto(s)
Ceramidas/farmacología , Neoplasias de la Próstata/patología , Animales , Apoptosis/efectos de los fármacos , Línea Celular Tumoral , Ceramidas/uso terapéutico , Ensayos de Selección de Medicamentos Antitumorales , Humanos , Masculino , Ratones , Ratones Endogámicos BALB C , Ratones Desnudos , Ratones SCID , Trasplante de Neoplasias , Neoplasias de la Próstata/tratamiento farmacológico , Ensayos Antitumor por Modelo de Xenoinjerto
18.
Mol Pharm ; 6(6): 1883-90, 2009.
Artículo en Inglés | MEDLINE | ID: mdl-19722701

RESUMEN

Tetraphenylphosphonium (TPP), a phosphonium cation, is a promising means for tumor imaging. A major contributor to the pharmacokinetics of phosphonium cations is the efflux transporter P-glycoprotein (P-gp). For this application it is important to ascertain the influence of the multidrug resistance system on TPP. Therefore, our aim was to characterize the interaction of TPP with P-gp, in vitro and in in vivo models. P-gp-mediated transport of [3H]-TPP was assessed in Caco-2 cells and ex vivo in rat intestinal wall by the use of a diffusion cell system. The distribution of [3H]-TPP across the blood-brain barrier (BBB) was studied in rats and mice treated with P-gp modulators and in Mdr-1a/b((-/-)) knockout mice. The in vitro permeability coefficient of basolateral-to-apical transfer (PappB-A) of [3H]-TPP was 8-fold greater than apical-to-basolateral (PappA-B) coefficient, indicative of net mucosal secretion. A concentration dependent decrease of this secretion was obtained by the P-gp substrate verapamil, while no effect was evident by the MRP2 inhibitor MK-571 and the BCRP inhibitor FTC. [3H]-TPP transfer across rat jejunum wall was directional and concentration-dependent. 2,4-Dinitrophenol, cyclosporin A (CsA), verapamil and PSC-833 enhanced A-B transport of TPP 3.6-fold, 4-fold, 4.6-fold and 5.3-fold respectively. Likewise, PappA-B of [3H]-TPP was 5-fold greater in P-gp knockout mice than in controls. In vivo, PSC-833, P-gp inhibitor, significantly increased the uptake of [3H]-TPP in the liver, heart, small intestine and the lungs but not the brain. Similar results were obtained in P-gp knockout mice. Our study demonstrates that P-gp mediates TPP efflux in vitro and in vivo; however, the consistently poor BBB permeation of TPP in all in vivo studies including P-gp knockout animals indicates that it is most likely mediated by other mechanisms. These findings are important for optimized clinical application of TPP as an imaging agent in cancer.


Asunto(s)
Miembro 1 de la Subfamilia B de Casetes de Unión a ATP/metabolismo , Barrera Hematoencefálica/metabolismo , Mucosa Intestinal/metabolismo , Compuestos Onio/metabolismo , Compuestos Organofosforados/metabolismo , 2,4-Dinitrofenol/farmacología , Miembro 1 de la Subfamilia B de Casetes de Unión a ATP/antagonistas & inhibidores , Miembro 1 de la Subfamilia B de Casetes de Unión a ATP/genética , Miembro 1 de la Subfamilia B de Casetes de Unión a ATP/fisiología , Transportador de Casetes de Unión a ATP, Subfamilia G, Miembro 2 , Transportadoras de Casetes de Unión a ATP/antagonistas & inhibidores , Transportadoras de Casetes de Unión a ATP/metabolismo , Animales , Transporte Biológico/efectos de los fármacos , Células CACO-2 , Bloqueadores de los Canales de Calcio/farmacología , Ciclosporina/farmacología , Ciclosporinas/farmacología , Inhibidores Enzimáticos/farmacología , Femenino , Humanos , Técnicas In Vitro , Antagonistas de Leucotrieno/farmacología , Ratones , Ratones Noqueados , Proteína 2 Asociada a Resistencia a Múltiples Medicamentos , Proteínas Asociadas a Resistencia a Múltiples Medicamentos/antagonistas & inhibidores , Proteínas Asociadas a Resistencia a Múltiples Medicamentos/metabolismo , Propionatos/farmacología , Quinolinas/farmacología , Ratas , Desacopladores/farmacología , Verapamilo/farmacología
19.
Diabetes Metab Res Rev ; 24(5): 413-21, 2008.
Artículo en Inglés | MEDLINE | ID: mdl-18561210

RESUMEN

BACKGROUND: Heparanase is an endo-beta-D-glucuronidase that cleaves heparan sulfate saccharide chains. The enzyme promotes cell adhesion, migration and invasion, and was shown to play a significant role in cancer metastasis and angiogenesis. METHODS: The present study focuses on the involvement of heparanase in autoimmunity, applying the murine non-obese diabetic (NOD) model, a T-cell-dependent disease often used to investigate the pathophysiology of type 1 diabetes. RESULTS: It was found that intra-peritoneal administration of heparanase ameliorated the clinical signs of the disease. In vitro studies revealed that heparanase has an inhibitory effect on the activation of T-cells through modulation of their repertoire of cytokines indicated by a marked increase in the levels of IL-4 and IL-10, and a parallel decrease in IL-12, tumour necrosis factor-alpha (TNF-alpha) and interferon-gamma (IFN-gamma). CONCLUSIONS: We suggest that heparanase induces a shift from a Th1- to Th2-phenotype, resulting in inhibition of diabetes in NOD mice and possibly other autoimmune disorders.


Asunto(s)
Citocinas/biosíntesis , Diabetes Mellitus Tipo 1/prevención & control , Glucuronidasa/farmacología , Activación de Linfocitos/efectos de los fármacos , Animales , Femenino , Humanos , Interferón gamma/biosíntesis , Interleucina-10/biosíntesis , Interleucina-12/biosíntesis , Interleucina-4/biosíntesis , Ratones , Ratones Endogámicos NOD , Células TH1/fisiología , Células Th2/fisiología , Factor de Necrosis Tumoral alfa
20.
Isr Med Assoc J ; 10(1): 20-5, 2008 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-18300565

RESUMEN

BACKGROUND: Standard CD44 and its alternatively spliced variants were found to be associated with the metastatic potential of tumor cells and with cell migration of autoimmune inflammatory cells, including cells involved in experimental insulin-dependent diabetes mellitus. OBJECTIVES: To investigate whether induction of anti-CD44 immune reactivity, through cDNA vaccination, could attenuate IDDM in a transfer model of NOD mice. METHODS: Our vaccination technique involved the insertion of CD44s or CD44v cDNA into a silicone tube filled with a 2.5 cm long segment of hydroxylated-polyvinyl acetate wound dressing sponge (forming a virtual lymph node) which was implanted under the skin of male NOD recipients reconstituted with diabetogenic spleen cells of female NOD donors. The VLN were implanted 20 days before and 3 days after cell transfer. RESULTS: In contrast to control groups of recipient mice, recipients vaccinated with VLN loaded with CD44v or CD44s cDNAs developed resistance to IDDM almost to the same extent. Our results suggest that the gene vaccination effect was mediated by anti-CD44 antibody rather than by cellular immunity. Histopathological examinations revealed a significant protection of pancreatic islets in the DNA-vaccinated recipients, whereas the islets of control recipients of diabetogenic cells were almost totally destroyed. CONCLUSIONS: These findings may open new opportunities for IDDM therapy in the future.


Asunto(s)
Diabetes Mellitus Tipo 1/tratamiento farmacológico , Receptores de Hialuranos/genética , Vacunas de ADN , Animales , Diabetes Mellitus Tipo 1/inmunología , Femenino , Receptores de Hialuranos/inmunología , Islotes Pancreáticos/metabolismo , Masculino , Ratones , Páncreas/metabolismo , Páncreas/patología , Vacunación/métodos
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA