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.
Neurosci Lett ; 607: 108-113, 2015 Oct 21.
Artigo em Inglês | MEDLINE | ID: mdl-26424077

RESUMO

Burn injuries have been identified as the primary cause of injury in 5% of U.S. military personnel evacuated from Operations Iraqi Freedom and Enduring Freedom. Severe burn-associated pain is typically treated with opioids such as fentanyl, morphine, and methadone. Side effects of opioids include respiratory depression, cardiac depression, decrease in motor and cognitive function, as well as the development of hyperalgesia, tolerance and dependence. These effects have led us to search for novel analgesics for the treatment of burn-associated pain in wounded combat service members. Tetrodotoxin (TTX) is a selective voltage-gated sodium channel blocker currently in clinical trials as an analgesic. A phase 3 clinical trial for cancer-related pain has been completed and phase 3 clinical trials on chemotherapy-induced neuropathic pain are planned. It has also been shown in mice to inhibit the development of chemotherapy-induced neuropathic pain. TTX was originally identified as a neurotoxin in marine animals but has now been shown to be safe in humans at therapeutic doses. The antinociceptive effects of TTX are thought to be due to inhibition of Na(+) ion influx required for initiation and conduction of nociceptive impulses. One TTX sensitive sodium channel, Nav1.7, has been shown to be essential in lowering the heat pain threshold after burn injuries. To date, the analgesic effect of TTX has not been tested in burn-associated pain. Male Sprague-Dawley rats were subjected to a full thickness thermal injury on the right hind paw. TTX (8 µg/kg) was administered once a day systemically by subcutaneous injection beginning 3 days post thermal injury and continued through 7 days post thermal injury. Thermal hyperalgesia and mechanical allodynia were assessed 60 and 120 min post injection on each day of TTX treatment. TTX significantly reduced thermal hyperalgesia at all days tested and had a less robust, but statistically significant suppressive effect on mechanical allodynia. These results suggest that systemic TTX may be an effective, rapidly acting analgesic for battlefield burn injuries and has the potential for replacing or reducing the need for opioid analgesics.


Assuntos
Analgésicos/uso terapêutico , Queimaduras/tratamento farmacológico , Hiperalgesia/tratamento farmacológico , Dor/tratamento farmacológico , Tetrodotoxina/uso terapêutico , Analgésicos Opioides/uso terapêutico , Animais , Queimaduras/fisiopatologia , Temperatura Alta , Hiperalgesia/fisiopatologia , Masculino , Morfina/uso terapêutico , Dor/fisiopatologia , Estimulação Física , Ratos Sprague-Dawley
2.
Cancer Res ; 75(17): 3636-49, 2015 Sep 01.
Artigo em Inglês | MEDLINE | ID: mdl-26141860

RESUMO

Metastasis is the main reason for lung cancer-related mortality, but little is known about specific determinants of successful dissemination from primary tumors and metastasis initiation. Here, we show that CD133(+)/CXCR4(+) cancer-initiating cells (CIC) directly isolated from patient-derived xenografts (PDX) of non-small cell lung cancer are endowed with superior ability to seed and initiate metastasis at distant organs. We additionally report that CXCR4 inhibition successfully prevents the increase of cisplatin-resistant CD133(+)/CXCR4(+) cells in residual tumors and their metastatization. Immunophenotypic analysis of lung tumor cells intravenously injected or spontaneously disseminated to murine lungs demonstrated the survival advantage and increased colonization ability of a specific subset of CD133(+)/CXCR4(+) with reduced expression of epithelial cell adhesion molecule (EpCAM(-)), which also shows the greatest in vitro invasive potential. We next prove that recovered disseminated cells from lungs of PDX-bearing mice enriched for CD133(+)/CXCR4(+)/EpCAM(-) CICs are highly tumorigenic and metastatic. Importantly, microenvironment stimuli eliciting epithelial-to-mesenchymal transition, including signals from cancer-associated fibroblasts, are able to increase the dissemination potential of lung cancer cells through the generation of the CD133(+)/CXCR4(+)/EpCAM(-) subset. These findings also have correlates in patient samples where disseminating CICs are enriched in metastatic lymph nodes (20-fold, P = 0.006) and their detection in primary tumors is correlated with poor clinical outcome (disease-free survival: P = 0.03; overall survival: P = 0.05). Overall, these results highlight the importance of specific cellular subsets in the metastatic process, the need for in-depth characterization of disseminating tumor cells, and the potential of therapeutic strategies targeting both primary tumor and tumor-microenvironment interactions.


Assuntos
Antígenos CD/biossíntese , Antígenos de Neoplasias/biossíntese , Carcinoma Pulmonar de Células não Pequenas/genética , Moléculas de Adesão Celular/biossíntese , Glicoproteínas/biossíntese , Neoplasias Pulmonares/genética , Receptores CXCR4/biossíntese , Antígeno AC133 , Idoso , Idoso de 80 Anos ou mais , Animais , Antígenos CD/genética , Antígenos de Neoplasias/genética , Carcinoma Pulmonar de Células não Pequenas/patologia , Moléculas de Adesão Celular/genética , Linhagem Celular Tumoral , Linhagem da Célula , Cisplatino/administração & dosagem , Intervalo Livre de Doença , Molécula de Adesão da Célula Epitelial , Transição Epitelial-Mesenquimal/efeitos dos fármacos , Feminino , Regulação Neoplásica da Expressão Gênica/efeitos dos fármacos , Glicoproteínas/genética , Humanos , Neoplasias Pulmonares/patologia , Masculino , Camundongos , Pessoa de Meia-Idade , Invasividade Neoplásica/genética , Células-Tronco Neoplásicas/patologia , Peptídeos/genética , Receptores CXCR4/genética , Transdução de Sinais/efeitos dos fármacos
3.
Wound Repair Regen ; 22(5): 622-30, 2014.
Artigo em Inglês | MEDLINE | ID: mdl-25139227

RESUMO

Effective prevention and treatment of hypertrophic scars (HTSs), a dermal form of fibrosis that frequently occurs following thermal injury to deep dermis, are unsolved significant clinical problems. Previously, we have found that stromal cell-derived factor 1/CXCR4 signaling is up-regulated during wound healing in burn patients and HTS tissue after thermal injury. We hypothesize that blood-borne mononuclear cells are recruited into wound sites after burn injury through the chemokine pathway of stromal cell-derived factor 1 and its receptor CXCR4. Deep dermal injuries to the skin are often accompanied by prolonged inflammation, which leads to chemotaxis of mononuclear cells into the wounds by chemokine signaling where fibroblast activation occurs and ultimately HTS are formed. Blocking mononuclear cell recruitment and fibroblast activation, CXCR4 antagonism is expected to reduce or minimize scar formation. In this study, the inhibitory effect of CXCR4 antagonist CTCE-9908 on dermal fibrosis was determined in vivo using a human HTS-like nude mouse model, in which split-thickness human skin is transplanted into full-thickness dorsal excisional wounds in athymic mice, where these wounds subsequently develop fibrotic scars that resemble human HTS as previously described. CTCE-9908 significantly attenuated scar formation and contraction, reduced the accumulation of macrophages and myofibroblasts, enhanced the remodeling of collagen fibers, and down-regulated the gene and protein expression of fibrotic growth factors in the human skin tissues. These findings support the potential therapeutic value of CXCR4 antagonist in dermal fibrosis and possibly other fibroproliferative disorders.


Assuntos
Cicatriz Hipertrófica/prevenção & controle , Derme/efeitos dos fármacos , Peptídeos/farmacologia , Receptores CXCR4/antagonistas & inibidores , Cicatrização/efeitos dos fármacos , Adulto , Animais , Cicatriz Hipertrófica/patologia , Derme/patologia , Modelos Animais de Doenças , Feminino , Fibrose , Humanos , Masculino , Camundongos Nus , Pessoa de Meia-Idade , Transplante de Pele
4.
BMC Urol ; 14: 12, 2014 Jan 28.
Artigo em Inglês | MEDLINE | ID: mdl-24472670

RESUMO

BACKGROUND: CXCL12/CXCR4 transactivation of epidermal growth factor family receptors in lipid raft membrane microdomains on cell surface is thought to mediate tumor growth and subsequent development of metastatic disease. CTCE-9908 is a known inhibitor of CXCR4. Herein, we tested the efficacy of CTCE-9908 in inhibiting prostate cancer cell growth, invasion, and metastasis. METHODS: We used a panel of in vitro assays utilizing human prostate cancer cell lines and an in vivo orthotopic prostate cancer model to assess the anti-tumoral activity of CTCE-9908. RESULTS: We demonstrated that (a) CTCE-9908 treatment resulted in no significant change in the growth of PC-3 and C4-2B cells; (b) 50 µg/ml of CTCE-9908 inhibited the invasive properties of PC-3 cells; (c) 25 mg/kg of CTCE-9908 did not alter primary tumor growth but it did significantly reduce total tumor burden in the animal including the growth of prostate and soft tissue metastases to lymph node and distant organ tissues. Histological analysis showed that CTCE-9908 treatment resulted in tumor necrosis in primary prostate tumors and no significant change in proliferation of tumor cells as measured by Ki-67 staining; (d) CTCE-9908 inhibited the tumor angiogenesis as measured by CD34 positive vessels in tumors. CONCLUSIONS: These data suggest that CXCR4 inhibition by CTCE-9908 decreases the invasion potential in vitro, which then translated to a reduction of tumor spread with associated reduction in angiogenesis. Hence, CTCE-9908 may prove to be an efficacious novel agent to prevent and treat the spread of metastatic prostate cancer.


Assuntos
Terapia de Alvo Molecular/métodos , Peptídeos/administração & dosagem , Neoplasias da Próstata/tratamento farmacológico , Neoplasias da Próstata/secundário , Receptores CXCR4/antagonistas & inibidores , Antineoplásicos/administração & dosagem , Linhagem Celular Tumoral , Sobrevivência Celular/efeitos dos fármacos , Humanos , Masculino , Neoplasias da Próstata/fisiopatologia , Receptores CXCR4/metabolismo , Resultado do Tratamento
5.
Inflammation ; 35(1): 130-7, 2012 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-21274742

RESUMO

The chemokine CXC receptor 4 (CXCR4) is activated by stromal cell-derived factor (SDF-1α). CXCR4 may be part of a lipopolysaccharide (LPS) sensing co-clustering complex that modulates TLR4 activation and evidence suggest that SDF-1α can activate anti-inflammatory signaling pathways and suppress inflammation. In the present study we examined the hypothesis that the SDF-1α peptide analog and CXCR4 agonist CTCE-0214 is anti-inflammatory in three distinct models of murine systemic inflammation. Our findings demonstrate that CTCE-0214 in vivo significantly suppressed plasma tumor necrosis factor alpha (TNF-α) increases in acute endotoxemia and following zymosan-induced multiple organ dysfunction syndrome (MODS). In both models, CTCE-0214 did not suppress plasma increases in the anti-inflammatory cytokine interleukin (IL)-10. CTCE-0214 improved survival without antibiotics in a model of severe sepsis induced by cecal ligation and puncture (CLP). CTCE-0214 also decreased plasma increases in IL-6 but not TNF-α and IL-10 in response to CLP-induced inflammation. We demonstrated in a moderately severe model of CLP (one puncture) that IL-6 levels at 24 h were similar to sham controls. However in severe CLP (two punctures) plasma IL-6 levels were markedly elevated. Plasma SDF-1α levels varied inversely with the plasma IL-6. In addition to the beneficial effect of CTCE-0214 in these models of systemic inflammation in vivo, we also demonstrated that the analog dose dependently suppressed LPS-induced IL-6 production in bone marrow-derived macrophages. CTCE-0214 therefore may be beneficial in controlling inflammation sepsis and systemic inflammatory syndromes.


Assuntos
Quimiocina CXCL12/metabolismo , Insuficiência de Múltiplos Órgãos/tratamento farmacológico , Receptores CXCR4/agonistas , Receptores CXCR4/imunologia , Síndrome de Resposta Inflamatória Sistêmica/tratamento farmacológico , Animais , Quimiocina CXCL12/sangue , Quimiocina CXCL12/farmacologia , Modelos Animais de Doenças , Endotoxemia/patologia , Interleucina-10/biossíntese , Interleucina-10/sangue , Interleucina-6/sangue , Lipopolissacarídeos/imunologia , Macrófagos/efeitos dos fármacos , Macrófagos/metabolismo , Masculino , Camundongos , Insuficiência de Múltiplos Órgãos/induzido quimicamente , Sepse/tratamento farmacológico , Fator de Necrose Tumoral alfa/sangue , Zimosan
6.
J Heart Lung Transplant ; 30(8): 955-62, 2011 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-21620738

RESUMO

BACKGROUND: Vascular endothelial growth factor (VEGF)-D is a member of the VEGF family, which can induce angiogenesis and lymphangiogenesis. We have previously demonstrated a role for VEGF-A in cardiac allograft vasculopathy (CAV). Our experiments profile the expression and localization of VEGF-D in human native atherosclerosis (NA), diabetes mellitus with atherosclerosis (DM) and CAV, and we investigate its ability to induce low-density lipoprotein (LDL) permeability in human cardiac microvascular endothelial cells (HCMEC). METHODS: VEGF-D mRNA and protein expression was characterized in coronary arteries and intramyocardial arterioles in NA, DM and CAV using in situ hybridization and immunohistochemical staining. Transendothelial electrical resistance (TER) measurements and immunocytochemical staining for platelet and endothelial cell adhesion molecule-1 and zonula occludens-1 were used to assess endothelial barrier and tight junctional integrity. LDL permeability in response to treatment with VEGF-D was measured using fluorometry in confluent HCMEC. RESULTS: Image quantitation demonstrated significant increases in VEGF-D immunoreactivity in the media of coronary arteries and intramyocardial arterioles of CAV cases, and in the intima and media of coronary arteries of DM cases. Treatment with VEGF-D, in vitro, significantly increased LDL passage through HCMEC monolayers. In conjunction, treatment with VEGF-D significantly decreased TER measurements 2 hours post-treatment and induced the formation of intercellular gaps through an extracellular signal-regulated kinase 1/2 (ERK1/2)-dependent pathway. CONCLUSIONS: VEGF-D is overexpressed in the arteries of CAV and DM cases. Treatment with VEGF-D can disrupt HCMEC tight junctions, resulting in the formation of intercellular gaps, and can also significantly increase LDL permeability through confluent monolayers.


Assuntos
Aterosclerose/metabolismo , Permeabilidade da Membrana Celular/fisiologia , Doença da Artéria Coronariana/metabolismo , Angiopatias Diabéticas/metabolismo , Endotélio Vascular/fisiopatologia , Lipoproteínas LDL/metabolismo , Fator D de Crescimento do Endotélio Vascular/metabolismo , Adolescente , Adulto , Arteríolas/metabolismo , Permeabilidade da Membrana Celular/efeitos dos fármacos , Células Cultivadas , Vasos Coronários/metabolismo , Endotélio Vascular/patologia , Feminino , Humanos , Técnicas In Vitro , Masculino , Proteína Quinase 1 Ativada por Mitógeno/metabolismo , Proteína Quinase 3 Ativada por Mitógeno/metabolismo , Junções Íntimas/fisiologia , Túnica Íntima/metabolismo , Túnica Média/metabolismo , Fator D de Crescimento do Endotélio Vascular/farmacologia , Adulto Jovem
7.
J Heart Lung Transplant ; 28(9): 950-7, 2009 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-19716049

RESUMO

BACKGROUND: Endothelial cell hyperpermeability is a proposed mechanism of increased lipid insudation into the vessel walls of allografts. Vascular endothelial growth factor (VEGF) is a potent inducer of vascular permeability and its expression is upregulated in human heart allografts. The goal of these experiments was to investigate the effects of VEGF on low-density lipoprotein (LDL) permeability through confluent monolayers of human cardiac microvascular endothelial cells (HCMEC) in vitro. METHODS: VEGF mRNA and protein expression was characterized in coronary arteries from cardiac allograft vasculopathy patients as compared with healthy controls using in situ hybridization and immunohistochemical staining of sub-adjacent sections. HCMEC were grown to confluence and treated with VEGF-A(121) or VEGF-A(165). Permeability of LDL in confluent endothelial monolayers was measured using fluorometry. Transendothelial electrical resistance (TER) measurements were used to indirectly measure the tight junctional status. Immunocytochemical staining was performed to visualize changes in CD31 and zonula occludens-1. RESULTS: We observed significant increases in VEGF expression within the superficial and deep intima and media of coronaries from allografts, as compared with controls. In vitro treatment with VEGF-A(121) and VEGF-A(165) significantly increased LDL passage through endothelial monolayers. We further showed that VEGF-A(121) and VEGF-A(165) caused significant decreases in TER at 2 to 4 hours post-treatment. Also, VEGF induced disruption of tight junctions, resulting in an increase in the intercellular gap formation. CONCLUSIONS: These results demonstrate that VEGF increases low-density lipoprotein permeability through endothelial monolayers, and this effect is correlated with VEGF-induced disruption of endothelial tight junctions resulting in the formation of intercellular gaps.


Assuntos
Permeabilidade da Membrana Celular/fisiologia , Circulação Coronária/fisiologia , Endotélio Vascular/fisiologia , Lipoproteínas LDL/fisiologia , Fragmentos de Peptídeos/farmacologia , Fator A de Crescimento do Endotélio Vascular/fisiologia , Adolescente , Adulto , Análise de Variância , Permeabilidade da Membrana Celular/efeitos dos fármacos , Circulação Coronária/efeitos dos fármacos , Endotélio Vascular/efeitos dos fármacos , Feminino , Transplante de Coração/fisiologia , Humanos , Imuno-Histoquímica , Hibridização In Situ , Masculino , Microcirculação/efeitos dos fármacos , Microcirculação/fisiologia , Fator A de Crescimento do Endotélio Vascular/genética , Fator A de Crescimento do Endotélio Vascular/farmacologia , Ferimentos e Lesões/mortalidade , Adulto Jovem
8.
Mol Cancer Ther ; 8(7): 1893-905, 2009 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-19567818

RESUMO

The chemokine receptor CXCR4 is expressed by malignant cells in ovarian cancer and is implicated in their growth and spread. We report here a unique mechanism of action of a small peptide antagonist of CXCR4 on ovarian cancer cells: induction of cell death by mitotic catastrophe. CTCE-9908 inhibited ovarian cancer cell migration to CXCL12, but on longer incubation, caused cell death in CXCR4-positive cells. CTCE-9908 did not cause apoptosis or cellular senescence, but induced multinucleation, G(2)-M arrest, and abnormal mitosis in ovarian cancer cells. This suggests that cell death was caused by mitotic catastrophe. Using microarray and Western blot analysis, we showed that CTCE-9908 deregulated DNA damage checkpoint proteins and spindle assembly checkpoint proteins at G(2)-M phases of the cell cycle. Combination treatment of CTCE-9908 and the drug paclitaxel led to an additive cytotoxicity that also involved mitotic catastrophe. We conclude that CTCE-9908 has a unique mechanism of action in ovarian cancer cells that seems to be CXCR4 specific.


Assuntos
Núcleo Celular/patologia , Mitose/efeitos dos fármacos , Neoplasias Ovarianas/tratamento farmacológico , Neoplasias Ovarianas/patologia , Peptídeos/farmacologia , Receptores CXCR4/antagonistas & inibidores , Antineoplásicos Fitogênicos/farmacologia , Apoptose/efeitos dos fármacos , Divisão Celular/efeitos dos fármacos , Movimento Celular/efeitos dos fármacos , Proliferação de Células/efeitos dos fármacos , Senescência Celular/efeitos dos fármacos , Dano ao DNA/efeitos dos fármacos , Replicação do DNA/efeitos dos fármacos , Quimioterapia Combinada , Feminino , Citometria de Fluxo , Fase G2/efeitos dos fármacos , Humanos , Neoplasias Ovarianas/metabolismo , Paclitaxel/farmacologia , Receptores CXCR4/metabolismo , Células Tumorais Cultivadas
9.
Prostate ; 69(13): 1460-9, 2009 Sep 15.
Artigo em Inglês | MEDLINE | ID: mdl-19588526

RESUMO

BACKGROUND: Recent reports have linked the survival-promoting effect of CXCR4 to the up regulation of Bcl-2 protein expression. MATERIALS AND METHODS: To further elucidate the relationship between Bcl-2 and CXCR4, tumorigenicity was evaluated in in vitro and in vivo models following treatment with CTCE-9908, a CXCR4 antagonist peptide. RESULTS: In vitro, CTCE-9908 inhibited cellular proliferation in PC-3-Bcl-2 and PC-3-Neo cell lines Furthermore in our xenograft model, CTCE-9908 delivered via daily intraperitoneal injections resulted in a statistically significant reduction in tumor size compared to control (396 + 205 mm(3) vs. 1,010 + 215 mm(3) respectively, p < 0.05) in the Bcl-2 expressing tumors. This reduction was associated with knockdown of VEGF, inhibition of angiogenesis and lymphangiogenesis, and induction of apoptosis. CTCE-9908 therapy was also associated with a marked reduction in intra-tumoral host cells expressing VEGFR1 and CD11b myeloid-derived suppressor cells (MDSC). CONCLUSION: These data show that CXCR4 antagonists represent a valuable addition to the cancer therapeutic arsenal. Such agents may have beneficial synergistic dual-effects in reducing tumor cell proliferation directly, and indirectly through perturbation of the tumor microenvironment. Further studies of the novel CTCE-9908 compound in prostate and other solid tumor inhibition are warranted. Prostate 69: 1460-1469, 2009. (c) 2009 Wiley-Liss, Inc.


Assuntos
Antineoplásicos/farmacologia , Peptídeos/farmacologia , Neoplasias da Próstata/tratamento farmacológico , Receptores CXCR4/antagonistas & inibidores , Animais , Antígeno CD11b/metabolismo , Divisão Celular/efeitos dos fármacos , Linhagem Celular Tumoral , Regulação para Baixo/efeitos dos fármacos , Humanos , Masculino , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Nus , Neoplasias da Próstata/patologia , Proteínas Proto-Oncogênicas c-akt/metabolismo , Receptor 1 de Fatores de Crescimento do Endotélio Vascular/metabolismo , Ensaios Antitumorais Modelo de Xenoenxerto
10.
Clin Biochem ; 42(10-11): 1162-5, 2009 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-19389390

RESUMO

OBJECTIVE: Can biochip arrays identify which individuals with metastatic disease will respond to an anti-metastatic agent? DESIGN AND METHODS: Cytokine and cell adhesion arrays (Randox Ltd) were measured over 1 month in 9 research participants receiving CTCE-9908 in a Phase I/II study. RESULTS: Research participants with stable disease (n=2) had significantly higher soluble VCAM-1 as compared to those that progressed. DISCUSSION: VCAM-1 measurement early during CTCE-9908 treatment might be used as a surrogate for response.


Assuntos
Antineoplásicos/uso terapêutico , Biomarcadores Tumorais/análise , Metástase Neoplásica/tratamento farmacológico , Peptídeos/uso terapêutico , Análise Serial de Proteínas/métodos , Adulto , Idoso , Moléculas de Adesão Celular/sangue , Citocinas/sangue , Demografia , Ensaios de Seleção de Medicamentos Antitumorais , Feminino , Humanos , Masculino , Pessoa de Meia-Idade
11.
J Immunol ; 182(7): 4448-58, 2009 Apr 01.
Artigo em Inglês | MEDLINE | ID: mdl-19299746

RESUMO

Among various surface molecules screened, CXCR4 was significantly up-regulated on monocytes, neutrophils, B cell subsets, and plasma cells in multiple murine models of lupus with active nephritis, including B6.Sle1Yaa, BXSB, and MRL.lpr. TLR-mediated signaling and inflammatory cytokines accounted in part for this increase. Increased CXCR4 expression was associated with functional consequences, including increased migration and enhanced B cell survival. Simultaneously, the ligand for CXCR4, CXCL12, was significantly up-regulated in the nephritic kidneys. Treatment with a peptide antagonist of CXCR4 prolonged survival and reduced serum autoantibodies, splenomegaly, intrarenal leukocyte trafficking, and end organ disease in a murine model of lupus. These findings underscore the pathogenic role of CXCR4/CXCL12 in lymphoproliferative lupus and lupus nephritis and highlight this axis as a promising therapeutic target in this disease.


Assuntos
Quimiocina CXCL12/biossíntese , Leucócitos/imunologia , Lúpus Eritematoso Sistêmico/imunologia , Receptores CXCR4/biossíntese , Animais , Quimiocina CXCL12/imunologia , Quimiotaxia de Leucócito/imunologia , Citocinas/imunologia , Ensaio de Imunoadsorção Enzimática , Feminino , Citometria de Fluxo , Imuno-Histoquímica , Inflamação/etiologia , Inflamação/imunologia , Inflamação/metabolismo , Leucócitos/metabolismo , Lúpus Eritematoso Sistêmico/complicações , Nefrite Lúpica/etiologia , Nefrite Lúpica/imunologia , Nefrite Lúpica/patologia , Masculino , Camundongos , Receptores CXCR4/imunologia , Receptores Toll-Like/imunologia , Receptores Toll-Like/metabolismo , Regulação para Cima
12.
Oncol Rep ; 21(3): 761-7, 2009 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-19212637

RESUMO

Metastasis occurs, in part, due to tumor cell responses to chemokine secretion by ectopic organs or tissues. SDF-1 is constitutively expressed in tissues where metastases frequently develop while breast carcinoma cells express the receptor for SDF-1, CXCR4, which is correlated with increased bone metastasis and poor overall survival. We hypothesized that treatment with a CXCR4 antagonist, CTCE-9908, would decrease incidence of bone and lung metastasis. Treatment with CTCE-9908 (25 mg/kg) began the day prior to or the day of intravenous or intracardiac tumor cell inoculation of MDA-MB-231 human breast carcinoma cells expressing enhanced green fluorescent protein (GFP) into athymic mice. After 5 or 8 weeks (i.c. and i.v. injections, respectively), the presence of fluorescent foci at metastatic sites was assessed. Somewhat surprisingly, CTCE-9908 treatment did not decrease incidence of metastasis as hypothesized. However, CTCE-9908 did decrease metastatic burden (i.e., size of metastases) in all organs examined (lungs, bone, heart, liver, kidneys, pancreas and spleen). Based upon this and other studies, the use of CTCE-9908 is promising as an adjuvant therapy for metastatic disease.


Assuntos
Antineoplásicos/uso terapêutico , Neoplasias Ósseas/prevenção & controle , Neoplasias Pulmonares/prevenção & controle , Neoplasias Mamárias Experimentais/tratamento farmacológico , Neoplasias Mamárias Experimentais/patologia , Peptídeos/uso terapêutico , Receptores CXCR4/antagonistas & inibidores , Animais , Neoplasias Ósseas/secundário , Feminino , Humanos , Neoplasias Pulmonares/secundário , Camundongos , Camundongos Nus , Receptores CXCR4/efeitos dos fármacos , Ensaios Antitumorais Modelo de Xenoenxerto
13.
Clin Cancer Res ; 14(24): 7975-80, 2008 Dec 15.
Artigo em Inglês | MEDLINE | ID: mdl-19088012

RESUMO

The majority of current cancer therapies focus on a primary tumor approach. However, it is metastases that cause the majority of cancer deaths. The metastatic process has been shown repeatedly to be greatly influenced by chemokines such as CXCL12 [stromal cell derived factor-1 (SDF-1)] and its receptor CXCR4. The activation of this pathway has been reported to modulate cell migration, survival, proliferation, and gene transcription through G proteins, phosphoinositide-3 kinase, Akt, extracellular signal-regulated kinase, arrestin, and Janus-activated kinase/signal transducers and activators of transcription. A wide variety of strategies, such as peptides, small molecules, antibodies, and small interfering RNA, have been used to target this pathway. Treatments in combination with current therapies seem to be especially promising in preclinical studies. A few compounds are advancing into early stages of clinical development. In this article, we will review the development of CXCR4 antagonists in oncology.


Assuntos
Receptores CXCR4/antagonistas & inibidores , Quimiocina CXCL12/fisiologia , Humanos , Receptores CXCR4/fisiologia , Transdução de Sinais
14.
Clin Exp Metastasis ; 25(3): 201-11, 2008.
Artigo em Inglês | MEDLINE | ID: mdl-18071913

RESUMO

Metastasis continues to be the leading cause of mortality for patients with cancer. High expression of the chemokine receptor CXCR4 correlates with poor prognosis in many cancers, including osteosarcoma and melanoma. CXCL12, the ligand for CXCR4, is expressed at high levels in the lung and lymph node, which are the primary sites to which these tumors metastasize respectively. These findings suggest that therapy aimed at disruption of this specific receptor/ligand complex may lead to a decrease in metastases. CTCE-9908, a small peptide CXCR4 antagonist was utilized in two murine metastasis models to test this hypothesis. Treatment of osteosarcoma cells in vitro with CTCE-9908 led to the following changes: decreased adhesion, decreased migration, decreased invasion, and decreased growth rate. Following tail vein injection of osteosarcoma cells, mice that were treated with CTCE-9908 had a 50% reduction in the number of gross metastatic lung nodules and a marked decrease in micro-metastatic disease. Similar findings were observed following injection of melanoma cells and treatment with CTCE-9908. However, these results could only be consistently reproduced when the cells were pre-treated with the inhibitor. A novel ex vivo luciferase assay showed decreased numbers of cells in the lung immediately after injection into mice, when treated with CTCE-9908, suggesting the importance of interactions between the receptor and the ligand. Our findings show that inhibition of the CXCR4/CXCL12 pathway decreases metastatic disease in two murine tumor models and expands on previous reports to describe potential mechanisms of action.


Assuntos
Quimiocina CXCL12/antagonistas & inibidores , Neoplasias Pulmonares/prevenção & controle , Melanoma/prevenção & controle , Osteossarcoma/prevenção & controle , Peptídeos/uso terapêutico , Receptores CXCR4/antagonistas & inibidores , Animais , Western Blotting , Adesão Celular/efeitos dos fármacos , Adesão Celular/fisiologia , Movimento Celular , Proliferação de Células , Quimiocina CXCL12/metabolismo , Citoesqueleto , Feminino , Neoplasias Pulmonares/secundário , Metástase Linfática , Melanoma/secundário , Camundongos , Camundongos Endogâmicos BALB C , Invasividade Neoplásica , Osteossarcoma/secundário , Fragmentos de Peptídeos/farmacologia , RNA Mensageiro/genética , RNA Mensageiro/metabolismo , Receptores CXCR4/metabolismo , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Transdução de Sinais/efeitos dos fármacos , Células Tumorais Cultivadas
15.
J Biomed Biotechnol ; 2007(3): 26065, 2007.
Artigo em Inglês | MEDLINE | ID: mdl-17541466

RESUMO

Stromal cell-derived factor-1alpha (SDF-1alpha) has pleiotropic effects on hematopoietic progenitor cells (HPCs). We have monitored podia formation, migration, proliferation, and cell-cell adhesion of human HPC under the influence of SDF-1alpha, a peptide agonist of CXCR4 (CTCE-0214), a peptide antagonist (CTCE-9908), and a nonpeptide antagonist (AMD3100). Whereas SDF-1alpha induced migration of CD34(+) cells in a dose-dependent manner, CTCE-0214, CTCE-9908, and AMD3100 did not induce chemotaxis in this concentration range albeit the peptides CTCE-0214 and CTCE-9908 increased podia formation. Cell-cell adhesion of HPC to human mesenchymal stromal cells was impaired by the addition of SDF-1alpha, CTCE-0214, and AMD3100. Proliferation was not affected by SDF-1alpha or its analogs. Surface antigen detection of CXCR4 was reduced upon treatment with SDF-1alpha or AMD3100 and it was enhanced by CTCE-9908. Despite the fact that all these molecules target the same CXCR4 receptor, CXCR4 agonists and antagonists have selective effects on different functions of the natural molecule.

16.
J Neuroimmunol ; 184(1-2): 136-48, 2007 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-17291598

RESUMO

The mechanisms by which polymorphonuclear leukocytes (PMN) cross the human blood-brain barrier have not been fully elucidated. Using a well characterized in vitro model of the human BBB, we examined the role of endothelial cell adhesion molecules on the adhesion and transendothelial migration of PMN across primary cultures of human brain microvessel endothelial cells (HBMEC). A small number of PMN (0.06%) adhered to unstimulated HBMEC, and the basal adhesion was not affected by anti-adhesion molecule antibodies. Treatment of HBMEC with tumor necrosis factor (TNF)-alpha resulted in increased PMN adhesion that was significantly inhibited by blocking antibodies to E-selectin and ICAM-1, but not VCAM-1 or PECAM-1. A very small number of adherent PMN migrated across unstimulated HBMEC monolayers. Migration increased 2 to 20 fold following stimulation of HBMEC with TNF-alpha. Monoclonal antibody blocking studies showed that PMN used ICAM-1, but not VCAM-1, E-selectin or PECAM-1 to move across activated monolayers. Anti-adhesion molecule antibodies did not diminish the basal PMN migration. Ultrastructurally, PMN often aggregated on top and between adjacent endothelial cells and adhered by first extending pseudopodia along the apical endothelial surface. They then flattened and inserted themselves between endothelial cells in order to migrate across the monolayers. At the end of the migration period, the cultures resumed their continuity with no evidence of disruption. Transendothelial migration of PMN decreased the transendothelial electrical resistance and increased the permeability to horseradish peroxidase, which penetrated alongside the migrating leukocytes. A blocking antibody to ICAM-1 that greatly decreased migration, had no effect on the permeability changes. These studies provide insights into the mechanisms that regulate the entry of PMN into the brain and the increased permeability of the BBB in CNS inflammation.


Assuntos
Encéfalo/anatomia & histologia , Moléculas de Adesão Celular/fisiologia , Movimento Celular/fisiologia , Células Endoteliais/fisiologia , Endotélio Vascular/citologia , Neutrófilos/fisiologia , Análise de Variância , Anticorpos/farmacologia , Permeabilidade Capilar/efeitos dos fármacos , Permeabilidade Capilar/fisiologia , Adesão Celular/efeitos dos fármacos , Adesão Celular/fisiologia , Moléculas de Adesão Celular/imunologia , Movimento Celular/efeitos dos fármacos , Células Cultivadas , Células Endoteliais/efeitos dos fármacos , Células Endoteliais/ultraestrutura , Humanos , Microscopia Eletrônica/métodos , Fatores de Tempo , Fator de Necrose Tumoral alfa/farmacologia
17.
Stem Cells ; 24(1): 55-64, 2006 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-16123381

RESUMO

The SDF-1/CXCR4 axis has been implicated in the chemotaxis, homing, mobilization, and expansion of hematopoietic stem and progenitor cells. We studied the effects of a SDF-1 peptide analogue CTCE-0214 on the survival of cord blood CD34+ cells in culture, expansion, and engraftment of expanded cells in the nonobese diabetic/severe combined immunodeficient (NOD/SCID) mouse model. Our results demonstrated that CTCE-0214 synergized with thrombopoietin (TPO), stem cell factor (SCF), or flt-3 ligand (FL) on the survival of stem and progenitor cells in culture. Adding CTCE-0214 at a low concentration (0.01 ng/ml) for 4 days together with TPO, SCF, and FL significantly enhanced ex vivo expansion of CD34+ cells to subsets of primitive (CD34+CD38- cells, colony-forming unit-mixed [CFU-GEMMs]), erythroid (CFU-Es), myeloid (CFU-GMs), and megakaryocytic (CD61+CD41+ cells, CFU-MKs) progenitors, as well as their multilineage engraftment in NOD/SCID mice. Interestingly, the short exposure of expanded cells to CTCE-0214 (100 and 500 ng/ml) for 4 hours did not increase the quantity of progenitor cells but enhanced their engraftment capacity. The proportion of CD34+ cells expressing surface CXCR4 was decreased, but the overall number of this population increased upon expansion. The small peptide analogue of SDF-1 could be developed for ex vivo expansion and improving engraftment of cord blood transplantation.


Assuntos
Sobrevivência Celular/efeitos dos fármacos , Quimiocinas CXC/fisiologia , Citocinas/farmacologia , Proteínas de Membrana/farmacologia , Fator de Células-Tronco/farmacologia , Trombopoetina/farmacologia , Animais , Antígenos CD34/metabolismo , Quimiocina CXCL12 , Relação Dose-Resposta a Droga , Sinergismo Farmacológico , Sangue Fetal/citologia , Transplante de Células-Tronco Hematopoéticas , Células-Tronco Hematopoéticas , Camundongos , Camundongos SCID , Receptores CXCR4/metabolismo
18.
Eur J Pharmacol ; 514(2-3): 91-8, 2005 May 09.
Artigo em Inglês | MEDLINE | ID: mdl-15910796

RESUMO

The entry of lymphocytes into the brain is normally limited by the blood-brain barrier, however, during inflammation prominent lymphocytic infiltration occurs. In this study, we investigated the effects of nitric oxide (NO) on the adhesion of T cells to cultured human brain microvessel endothelial cells. T cell adhesion to unstimulated or tumor necrosis factor-alpha (TNF-alpha)-treated cells was quantified by counting the number of lymphocytes bound to the monolayer by light microscopy. TNF-alpha increased T cell adhesion in a time-dependent manner. Incubation of monolayers with NO donors decreased adhesion. This effect was blocked by a guanylyl cyclase inhibitor and mimicked by a cGMP agonist, and was thus dependent on the generation of cGMP. NO did not modulate adhesion molecule expression in the endothelial cells, suggesting an action on the T cells. Pre-treatment of T cells with NO or a cGMP agonist decreased binding to recombinant endothelial adhesion molecules. These findings suggest that NO can modulate the adhesion of T cells to human brain microvessel endothelial cells via a cGMP-dependent mechanism, and may thus regulate lymphocyte traffic during central nervous system inflammation.


Assuntos
GMP Cíclico/análogos & derivados , GMP Cíclico/metabolismo , Células Endoteliais/citologia , Óxido Nítrico/fisiologia , Transdução de Sinais/fisiologia , Linfócitos T/citologia , Encéfalo/irrigação sanguínea , Adesão Celular/efeitos dos fármacos , Adesão Celular/fisiologia , Células Cultivadas , GMP Cíclico/farmacologia , Relação Dose-Resposta a Droga , Selectina E/farmacologia , Inibidores Enzimáticos/farmacologia , Guanilato Ciclase/antagonistas & inibidores , Humanos , Molécula 1 de Adesão Intercelular/farmacologia , NG-Nitroarginina Metil Éster/farmacologia , Doadores de Óxido Nítrico/farmacologia , Óxido Nítrico Sintase/antagonistas & inibidores , Nitroprussiato/farmacologia , Compostos Nitrosos/farmacologia , Oxidiazóis/farmacologia , Molécula-1 de Adesão Celular Endotelial a Plaquetas/farmacologia , Quinoxalinas/farmacologia , Fator de Necrose Tumoral alfa/farmacologia , Molécula 1 de Adesão de Célula Vascular/farmacologia
19.
Exp Hematol ; 33(3): 295-307, 2005 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-15730853

RESUMO

OBJECTIVE: Mobilization of hematopoietic stem and progenitor cells (HSPC) by stromal cell-derived factor-1 (SDF-1) has been described; however, sustained adenoviral delivery or N-terminal modification was required for effect and could not be demonstrated with native protein. The aim of this study was to further investigate the SDF-1alpha/CXCR4 axis in HSPC mobilization using CTCE-0021, a cyclized CXCR4 agonist peptide, with comparable bioactivity and improved stability relative to SDF-1alpha. METHODS: Peripheral blood cells and hematopoietic progenitor cells (HPC) were quantitated in mice administered single or multiple doses of CTCE-0021 or SDF-1alpha, or mobilized by granulocyte colony-stimulating factor (G-CSF) in combination with CTCE-0021. Proteases, cytokines, and receptors implicated in HSPC mobilization were evaluated to determine mechanism of action. RESULTS: CTCE-0021 dose-dependently elevated blood neutrophils polymorphonuclear neutrophil [PMN] within 5 minutes that peaked after 1 hour and persisted for 24 hours. PMN mobilization could be maintained by daily dosing. CTCE-0021 mobilized colony-forming unit granulocyte macrophage (CFU-GM), burst-forming unit erythroid (BFU-E), and CFU-granulocyte-erythrocyte-monocyte-megakaryocyte (CFU-GEMM) that peaked within 1 hour after administration, and synergistically enhanced both PMN and HSPC mobilization when combined with G-CSF. Mobilization induced by CTCE-0021 was associated with rapid downregulation of CXCR4 expression on HPC. No appreciable changes in proteases implicated in HPC mobilization were observed. Significantly elevated plasma SDF-1 was detected in mobilized mice, which likely represents CTCE-0021. CONCLUSION: These studies indicate that CTCE-0021 is an efficient and rapid mobilizer of PMN and HPC when used alone and shows synergistic activity when used in combination with G-CSF. The mobilizing effect of this peptide appears to be mediated by downregulation of the CXCR4 receptor on HPC and altered chemokine gradient.


Assuntos
Quimiocinas CXC/administração & dosagem , Fator Estimulador de Colônias de Granulócitos/administração & dosagem , Mobilização de Células-Tronco Hematopoéticas , Peptídeos/administração & dosagem , Receptores CXCR4/agonistas , Animais , Quimiocina CXCL12 , Relação Dose-Resposta a Droga , Regulação para Baixo/efeitos dos fármacos , Sinergismo Farmacológico , Feminino , Células-Tronco Hematopoéticas/fisiologia , Injeções Subcutâneas , Camundongos , Camundongos Endogâmicos BALB C , Neutrófilos/fisiologia , Receptores CXCR4/metabolismo
20.
Biochem Biophys Res Commun ; 323(1): 142-8, 2004 Oct 08.
Artigo em Inglês | MEDLINE | ID: mdl-15351713

RESUMO

The hypothesis that the NO/cGMP pathway modulates PMN adhesion to human brain microvessel endothelial cells (HBMEC) was examined. Human PMN were incubated with resting or TNF-alpha-treated endothelial monolayers, and adhesion was quantified by light microscopy. TNF-alpha upregulated PMN adhesion in a time-dependent manner. Treatment of HBMEC with the NO donors SNP and DETA NONOate for 4 or 24 h decreased PMN adhesion. This was completely reversed by the guanylyl cyclase inhibitor ODQ, while addition of a cGMP agonist (8-Br-cGMP) decreased PMN adhesion. NO donors did not affect the levels of E-selectin or ICAM-1 in HBMEC. However, pre-treatment of PMN with NO donors or 8-Br-cGMP decreased their adhesion to recombinant E-selectin and ICAM-1, suggesting an effect of NO on PMN. These findings indicate that NO modulates PMN-HBMEC interactions through cGMP and decreases the binding of PMN to the adhesion molecules E-selectin and ICAM-1.


Assuntos
Encéfalo/irrigação sanguínea , Células Endoteliais/citologia , Endotélio Vascular/citologia , Neutrófilos/metabolismo , Óxido Nítrico/metabolismo , Western Blotting , Adesão Celular , Células Cultivadas , Proteínas Quinases Dependentes de GMP Cíclico/metabolismo , Selectina E/metabolismo , Humanos , Inflamação , Molécula 1 de Adesão Intercelular/metabolismo , Microcirculação , Modelos Biológicos , Compostos Nitrosos/farmacologia , Transdução de Sinais , Fatores de Tempo , Fator de Necrose Tumoral alfa/metabolismo , Regulação para Cima
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA