Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 21
Filtrar
Mais filtros











Base de dados
Intervalo de ano de publicação
1.
J Cancer Educ ; 2024 May 31.
Artigo em Inglês | MEDLINE | ID: mdl-38819526

RESUMO

Over the last two decades, strides in cancer prevention, earlier detection, and novel treatments have reduced overall cancer mortality; however, cancer health disparities (CHD) persist among demographically diverse and intersecting populations. The development of a culturally responsive workforce trained in interdisciplinary, team-based science is a key strategy for addressing these cancer disparities. The Cancer Research - Scholarship and Training Experience in Population Sciences (C-STEPS) program at the University of New Mexico Comprehensive Cancer Center is designed to increase and diversify the biomedical and behavioral research workforce by providing specialized and experiential curricula that highlight team-oriented cancer control and population science. Undergraduate students interested in CHD and in pursuing STEM-H (science, technology, engineering, mathematics, and health) graduate or professional degrees are eligible for the program. C-STEPS students are paired with a UNM faculty mentor, who guides the student's 10-week summer research experience. They receive mentorship and support from three layers-faculty, near-peers (graduate students), and peers (undergraduates who have completed the C-STEPS program previously). Students generate five products, including a capstone presentation, grounded in the research they conduct with their faculty mentors. Since its founding in 2021, C-STEPS has trained three cohorts with a total of 32 students. The C-STEPS program provides a unique team-science approach with multilayer mentoring to create a sustainable pipeline for the development of students interested in STEM-H fields and CHD research. The capstone project led to 47% of students presenting their work at conferences, and two publishing their manuscripts in peer-reviewed journals. Overall, 89% of students were either "satisfied" or "very satisfied" with the program and the same percentage recommended the program to other undergraduates.

2.
Trauma Surg Acute Care Open ; 9(Suppl 1): e001126, 2024.
Artigo em Inglês | MEDLINE | ID: mdl-38196934

RESUMO

Objectives: Trauma-induced coagulopathy (TIC) occurs in a subset of severely injured trauma patients. Despite having achieved surgical hemostasis, these individuals can have persistent bleeding, clotting, or both in conjunction with deranged coagulation parameters and typically require transfusion support with plasma, platelets, and/or cryoprecipitate. Due to the multifactorial nature of TIC, targeted interventions usually do not have significant clinical benefits. Therapeutic plasma exchange (TPE) is a non-specific modality of removing and replacing a patient's plasma in a euvolemic manner that can temporarily normalize coagulation parameters and remove deleterious substances, and may be beneficial in such patients with TIC. Methods: In a prospective case series, TPE was performed in severely injured trauma patients diagnosed with TIC and transfusion requirement. These individuals all underwent a series of at least 3 TPE procedures performed once daily with plasma as the exclusive replacement fluid. Demographic, injury, laboratory, TPE, and outcome data were collected and analyzed. Results: In total, 7 patients received 23 TPE procedures. All patients had marked improvements in routine coagulation parameters, platelet counts, a disintegrin and metalloproteinase with a thrombospondin type 1 motif, member 13 (ADAMTS13) activities, inflammatory markers including interleukin-6 concentrations, and organ system injuries after completion of their TPE treatments. All-cause mortality rates at 1 day, 7 days, and 30 days were 0%, 0%, and 43%, respectively, and all patients for whom TPE was initiated within 24 hours after injury survived to the 30-day timepoint. Surgical, critical care, and apheresis nursing personnel who were surveyed were universally positive about the utilization of TPE in this patient population. These procedures were tolerated well with the most common adverse event being laboratory-diagnosed hypocalcemia. Conclusion: TPE is feasible and tolerable in severely injured trauma patients with TIC. However, many questions remain regarding the application of TPE for these critically ill patients including identification of the optimal injured population, ideal time of treatment initiation, appropriate treatment intensity, and concurrent use of adjunctive treatments. Level of evidence: Level V.

3.
Cancers (Basel) ; 15(11)2023 May 26.
Artigo em Inglês | MEDLINE | ID: mdl-37296882

RESUMO

Dysregulated cellular processes drive malignant transformation, tumor progression, and metastasis, and affect responses to therapies [...].

4.
Stem Cell Reports ; 16(10): 2422-2431, 2021 10 12.
Artigo em Inglês | MEDLINE | ID: mdl-34534447

RESUMO

Hematopoietic stem and progenitor cell (HSPC) mobilization into the blood occurs under normal physiological conditions and is stimulated in the clinic to enable the isolation of HSPCs for transplantation therapies. In the present study, we identify the tetraspanin CD82 as a novel regulator of HSPC mobilization. Using a global CD82 knockout (CD82KO) mouse, we measure enhanced HSPC mobilization after granulocyte-colony stimulating factor (G-CSF) or AMD3100 treatment, which we find is promoted by increased surface expression of the sphingosine 1-phosphate receptor 1 (S1PR1) on CD82KO HSPCs. Additionally, we identify a disruption in S1PR1 internalization in CD82-deficient HSPCs, suggesting that CD82 plays a critical role in S1PR1 surface regulation. Finally, combining AMD3100 and anti-CD82 treatments, we detect enhanced mobilization of mouse HSPCs and human CD34+ cells in animal models. Together, these data provide evidence that CD82 is an important regulator of HSPC mobilization and suggests exploiting the CD82 scaffold as a therapeutic target to enhance stem cell isolation.


Assuntos
Mobilização de Células-Tronco Hematopoéticas , Células-Tronco Hematopoéticas/metabolismo , Proteína Kangai-1/fisiologia , Receptores de Esfingosina-1-Fosfato/metabolismo , Células-Tronco/metabolismo , Animais , Antígenos CD34/metabolismo , Regulação da Expressão Gênica , Fator Estimulador de Colônias de Granulócitos , Transplante de Células-Tronco Hematopoéticas , Humanos , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Transdução de Sinais , Tetraspaninas/fisiologia
5.
Cancers (Basel) ; 13(17)2021 Aug 26.
Artigo em Inglês | MEDLINE | ID: mdl-34503110

RESUMO

BACKGROUND: The development of non-small cell lung cancer (NSCLC) involves the progressive accumulation of genetic and epigenetic changes. These include somatic oncogenic KRAS and EGFR mutations and inactivating TP53 tumour suppressor mutations, leading to activation of canonical NF-κB. However, the mechanism(s) by which canonical NF-κB contributes to NSCLC is still under investigation. METHODS: Human NSCLC cells were used to knock-down RelA/p65 (RelA/p65KD) and investigate its impact on cell growth, and its mechanism of action by employing RNA-seq analysis, qPCR, immunoblotting, immunohistochemistry, immunofluorescence and functional assays. RESULTS: RelA/p65KD reduced the proliferation and tumour growth of human NSCLC cells grown in vivo as xenografts in immune-compromised mice. RNA-seq analysis identified canonical NF-κB targets mediating its tumour promoting function. RelA/p65KD resulted in the upregulation of the metastasis suppressor CD82/KAI1/TSPAN27 and downregulation of the proto-oncogene ROS1, and LGR6 involved in Wnt/ß-catenin signalling. Immunohistochemical and bioinformatics analysis of human NSCLC samples showed that CD82 loss correlated with malignancy. RelA/p65KD suppressed cell migration and epithelial-to-mesenchymal cell transition (EMT), mediated, in part, by CD82/KAI1, through integrin-mediated signalling involving the mitogenic ERK, Akt1 and Rac1 proteins. CONCLUSIONS: Canonical NF-κB signalling promotes NSCLC, in part, by downregulating the metastasis suppressor CD82/KAI1 which inhibits cell migration, EMT and tumour growth.

6.
Int J Biochem Cell Biol ; 137: 106029, 2021 08.
Artigo em Inglês | MEDLINE | ID: mdl-34174403

RESUMO

Acute myeloid leukemia (AML) is characterized by the disruption of myeloid differentiation and accumulation of blast cells in the bone marrow. While AML patients respond favorably to induction chemotherapy, long-term outcomes remain poor due to a high rate of chemoresistance. Advances with targeted therapies, which can be used in combination with conventional chemotherapy, have expanded therapeutic options for patients. However, remission is often short-lived and followed by disease relapse and drug resistance. Therefore, there is a substantial need to improve treatment options by identifying novel molecular and cellular targets that regulate AML chemosensitivity. Membrane scaffolds such as the tetraspanin family of proteins often serve as signaling mediators, translating extracellular signaling cues into intracellular signaling cascades. In this review, we discuss the conventional and targeted treatment strategies for AML and review chemoresistance mechanisms with a focus on the tetraspanin family of membrane scaffold proteins.


Assuntos
Antineoplásicos/farmacologia , Resistencia a Medicamentos Antineoplásicos , Regulação Leucêmica da Expressão Gênica/efeitos dos fármacos , Leucemia Mieloide Aguda/tratamento farmacológico , Tetraspaninas/metabolismo , Animais , Humanos , Leucemia Mieloide Aguda/genética , Leucemia Mieloide Aguda/metabolismo , Leucemia Mieloide Aguda/patologia , Tetraspaninas/genética
7.
Front Cell Dev Biol ; 8: 598, 2020.
Artigo em Inglês | MEDLINE | ID: mdl-32754593

RESUMO

Hematopoietic stem and progenitor cells (HSPCs) are responsible for the development, maintenance, and regeneration of all the blood forming cells in the body, and as such, are critical for a number of patient therapies. For successful HSPC transplantation, stem cells must traffic through the blood and home to the bone marrow (BM) microenvironment or "niche," which is composed of soluble factors, matrix proteins, and supportive cells. HSPC adhesion to, and signaling with, cellular and extracellular components of the niche provide instructional cues to balance stem cell self-renewal and differentiation. In this review, we will explore the regulation of these stem cell properties with a focus on the tetraspanin family of membrane proteins. Tetraspanins are molecular scaffolds that uniquely function to distribute proteins into highly organized microdomains comprising adhesion, signaling, and adaptor proteins. As such, tetraspanins contribute to many aspects of cell physiology as mediators of cell adhesion, trafficking, and signaling. We will summarize the many reports that identify tetraspanins as markers of specific HSPC populations. Moreover, we will discuss the various studies establishing the functional importance of tetraspanins in the regulation of essential HSPC processes including quiescence, migration, and niche adhesion. When taken together, studies outlined in this review suggest that several tetraspanins may serve as potential targets to modulate HSPC interactions with the BM niche, ultimately impacting future HSPC therapies.

8.
Oncogene ; 39(19): 3910-3925, 2020 05.
Artigo em Inglês | MEDLINE | ID: mdl-32203165

RESUMO

A principal challenge in treating acute myeloid leukemia (AML) is chemotherapy refractory disease. As such, there remains a critical need to identify key regulators of chemotherapy resistance in AML. In this study, we demonstrate that the membrane scaffold, CD82, contributes to the chemoresistant phenotype of AML. Using an RNA-seq approach, we identified the increased expression of the tetraspanin family member, CD82, in response to the chemotherapeutic, daunorubicin. Analysis of the TARGET and BEAT AML databases identifies a correlation between CD82 expression and overall survival of AML patients. Moreover, using a combination of cell lines and patient samples, we find that CD82 overexpression results in significantly reduced cell death in response to chemotherapy. Investigation of the mechanism by which CD82 promotes AML survival in response to chemotherapy identified a crucial role for enhanced protein kinase c alpha (PKCα) signaling and downstream activation of the ß1 integrin. In addition, analysis of ß1 integrin clustering by super-resolution imaging demonstrates that CD82 expression promotes the formation of dense ß1 integrin membrane clusters. Lastly, evaluation of survival signaling following daunorubicin treatment identified robust activation of p38 mitogen-activated protein kinase (MAPK) downstream of PKCα and ß1 integrin signaling when CD82 is overexpressed. Together, these data propose a mechanism where CD82 promotes chemoresistance by increasing PKCα activation and downstream activation/clustering of ß1 integrin, leading to AML cell survival via activation of p38 MAPK. These observations suggest that the CD82-PKCα signaling axis may be a potential therapeutic target for attenuating chemoresistance signaling in AML.


Assuntos
Integrina beta1/genética , Proteína Kangai-1/genética , Leucemia Mieloide Aguda/tratamento farmacológico , Proteína Quinase C-alfa/genética , Adulto , Idoso , Daunorrubicina/efeitos adversos , Daunorrubicina/farmacologia , Resistencia a Medicamentos Antineoplásicos/genética , Feminino , Regulação Neoplásica da Expressão Gênica/efeitos dos fármacos , Células HL-60 , Humanos , Leucemia Mieloide Aguda/genética , Leucemia Mieloide Aguda/patologia , Masculino , Pessoa de Meia-Idade , RNA-Seq , Proteínas Quinases p38 Ativadas por Mitógeno/genética
9.
Cancers (Basel) ; 10(10)2018 Sep 27.
Artigo em Inglês | MEDLINE | ID: mdl-30261690

RESUMO

The tumor microenvironment for epithelial ovarian cancer is complex and rich in bioactive molecules that modulate cell-cell interactions and stimulate numerous signal transduction cascades. These signals ultimately modulate all aspects of tumor behavior including progression, metastasis and therapeutic response. Many of the signaling pathways converge on the small GTPase Ras-related C3 botulinum toxin substrate (Rac)1. In addition to regulating actin cytoskeleton remodeling necessary for tumor cell adhesion, migration and invasion, Rac1 through its downstream effectors, regulates cancer cell survival, tumor angiogenesis, phenotypic plasticity, quiescence, and resistance to therapeutics. In this review we discuss evidence for Rac1 activation within the ovarian tumor microenvironment, mechanisms of Rac1 dysregulation as they apply to ovarian cancer, and the potential benefits of targeting aberrant Rac1 activity in this disease. The potential for Rac1 contribution to extraperitoneal dissemination of ovarian cancer is addressed.

10.
Mol Biol Cell ; 29(24): 2946-2958, 2018 11 26.
Artigo em Inglês | MEDLINE | ID: mdl-30133344

RESUMO

Hematopoietic stem and progenitor cell (HSPC) transplantation represents a treatment option for patients with malignant and nonmalignant hematological diseases. Initial steps in transplantation involve the bone marrow homing and engraftment of peripheral blood-injected HSPCs. In recent work, we identified the tetraspanin CD82 as a potential regulator of HSPC homing to the bone marrow, although its mechanism remains unclear. In the present study, using a CD82 knockout (CD82KO) mouse model, we determined that CD82 modulates HSPC bone marrow maintenance, homing, and engraftment. Bone marrow characterization identified a significant decrease in the number of long-term hematopoietic stem cells in the CD82KO mice, which we linked to cell cycle activation and reduced stem cell quiescence. Additionally, we demonstrate that CD82 deficiency disrupts bone marrow homing and engraftment, with in vitro analysis identifying further defects in migration and cell spreading. Moreover, we find that the CD82KO HSPC homing defect is due at least in part to the hyperactivation of Rac1, as Rac1 inhibition rescues homing capacity. Together, these data provide evidence that CD82 is an important regulator of HSPC bone marrow maintenance, homing, and engraftment and suggest exploiting the CD82 scaffold as a therapeutic target for improved efficacy of stem cell transplants.


Assuntos
Células da Medula Óssea/citologia , Células-Tronco Hematopoéticas/citologia , Células-Tronco Hematopoéticas/metabolismo , Proteína Kangai-1/metabolismo , Animais , Células da Medula Óssea/metabolismo , Movimento Celular/fisiologia , Células Cultivadas , Modelos Animais de Doenças , Feminino , Transplante de Células-Tronco Hematopoéticas/métodos , Proteína Kangai-1/deficiência , Proteína Kangai-1/genética , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout
11.
BMC Hematol ; 17: 3, 2017.
Artigo em Inglês | MEDLINE | ID: mdl-28174663

RESUMO

BACKGROUND: Chronic lymphocytic leukemia (CLL) is the most common leukemia in Western countries. The frequency of symptomatic central nervous system (CNS) involvement is unknown but thought to be a rare phenomenon. Currently there are no known risk factors for CNS involvement. CASE PRESENTATION: We describe a clinically staged low-risk CLL case that presented with symptomatic CNS involvement and progressed rapidly to death. Evaluation of the surface adhesion molecules identified a markedly altered expression pattern of the integrin, CD49d, and the tetraspanin, CD82, in the index case when compared to similar low-risk CLL cases. We found that the early Rai clinical stage CLL patients showed linear correlation for the co-expression of CD82 and CD49d. In contrast, this unique index case with CNS involvement, which has the same Rai clinical stage, had a significantly lower expression of CD82 and higher expression of CD49d. CONCLUSIONS: These data suggest that the expression profile of CD49d and CD82 may represent potential biomarkers for patients with increased propensity of CNS involvement. Moreover, this study illustrates the critical need for a better mechanistic understanding of how specific adhesion proteins regulate the interactions between CLL cells and various tissue sites.

12.
Sci Rep ; 6: 29859, 2016 07 15.
Artigo em Inglês | MEDLINE | ID: mdl-27417454

RESUMO

Patients with acute myeloid leukemia (AML) have increased myeloid cells within their bone marrow that exhibit aberrant signaling. Therefore, therapeutic targets that modulate disrupted signaling cascades are of significant interest. In this study, we demonstrate that the tetraspanin membrane scaffold, CD82, regulates protein kinase c alpha (PKCα)-mediated signaling critical for AML progression. Utilizing a palmitoylation mutant form of CD82 with disrupted membrane organization, we find that the CD82 scaffold controls PKCα expression and activation. Combining single molecule and ensemble imaging measurements, we determine that CD82 stabilizes PKCα activation at the membrane and regulates the size of PKCα membrane clusters. Further evaluation of downstream effector signaling identified robust and sustained activation of ERK1/2 upon CD82 overexpression that results in enhanced AML colony formation. Together, these data propose a mechanism where CD82 membrane organization regulates sustained PKCα signaling that results in an aggressive leukemia phenotype. These observations suggest that the CD82 scaffold may be a potential therapeutic target for attenuating aberrant signal transduction in AML.


Assuntos
Proteína Kangai-1/genética , Leucemia Mieloide Aguda/genética , Proteína Quinase C-alfa/genética , Tetraspaninas/genética , Medula Óssea/metabolismo , Medula Óssea/patologia , Linhagem Celular Tumoral , Progressão da Doença , Regulação Leucêmica da Expressão Gênica/genética , Humanos , Leucemia Mieloide Aguda/patologia , Lipoilação , Transdução de Sinais/genética , Tetraspaninas/metabolismo
13.
Mol Biol Cell ; 25(10): 1560-73, 2014 May.
Artigo em Inglês | MEDLINE | ID: mdl-24623721

RESUMO

Hematopoietic stem/progenitor cell (HSPC) interactions with the bone marrow microenvironment are important for maintaining HSPC self-renewal and differentiation. In recent work, we identified the tetraspanin protein, CD82, as a regulator of HPSC adhesion and homing to the bone marrow, although the mechanism by which CD82 mediated adhesion was unclear. In the present study, we determine that CD82 expression alters cell-matrix adhesion, as well as integrin surface expression. By combining the superresolution microscopy imaging technique, direct stochastic optical reconstruction microscopy, with protein clustering algorithms, we identify a critical role for CD82 in regulating the membrane organization of α4 integrin subunits. Our data demonstrate that CD82 overexpression increases the molecular density of α4 within membrane clusters, thereby increasing cellular adhesion. Furthermore, we find that the tight packing of α4 into membrane clusters depend on CD82 palmitoylation and the presence of α4 integrin ligands. In combination, these results provide unique quantifiable evidence of CD82's contribution to the spatial arrangement of integrins within the plasma membrane and suggest that regulation of integrin density by tetraspanins is a critical component of cell adhesion.


Assuntos
Adesão Celular/fisiologia , Células-Tronco Hematopoéticas/metabolismo , Integrina alfa4/metabolismo , Integrina alfa4beta1/metabolismo , Proteína Kangai-1/metabolismo , Adesão Celular/genética , Linhagem Celular , Membrana Celular/metabolismo , Movimento Celular , Junções Célula-Matriz/metabolismo , Estruturas Celulares/metabolismo , Endocitose , Fibronectinas/metabolismo , Humanos , Integrina alfa4/biossíntese , Integrina alfa4beta1/biossíntese , Proteína Kangai-1/biossíntese , Proteína Kangai-1/genética , Lipoilação , Transporte Proteico , Interferência de RNA , RNA Interferente Pequeno , Transdução de Sinais/fisiologia
14.
Methods Enzymol ; 534: 207-21, 2014.
Artigo em Inglês | MEDLINE | ID: mdl-24359956

RESUMO

Cell-cell communication is essential for an abundance of physiological processes. As such, various mechanisms have evolved to regulate and ensure proper cell-to-cell signaling. Recently, a novel mechanism of cell communication has emerged which involves the physical transfer of proteins, lipids, and nucleic acids between cells. Following this process termed intercellular transfer (ICT), the transferred molecules can signal within recipient cells by entering the endosomal system and trafficking to signaling endosomes. Signaling endosomes can modulate signal localization within the cell as well as the specificity of, and cross talk between, signaling pathways. As such, ICT into signaling endosomes has the potential to modify the signaling profile of the recipient cell. In this chapter, we describe the different methods of ICT as well as how transfer to signaling endosomes can be visualized and quantified.


Assuntos
Membrana Celular/metabolismo , Endossomos/metabolismo , Exossomos/metabolismo , Células-Tronco Hematopoéticas/metabolismo , Osteoblastos/metabolismo , Transporte Biológico , Comunicação Celular , Linhagem Celular , Membrana Celular/ultraestrutura , Endossomos/química , Exossomos/química , Citometria de Fluxo , Células-Tronco Hematopoéticas/citologia , Humanos , Metabolismo dos Lipídeos , Microscopia Confocal , Ácidos Nucleicos/metabolismo , Osteoblastos/citologia , Proteínas/metabolismo , Transdução de Sinais , Coloração e Rotulagem
15.
Blood ; 119(8): 1848-55, 2012 Feb 23.
Artigo em Inglês | MEDLINE | ID: mdl-22228628

RESUMO

Manipulation of hematopoietic stem/progenitor cells (HSPCs) ex vivo is of clinical importance for stem cell expansion and gene therapy applications. However, most cultured HSPCs are actively cycling, and show a homing and engraftment defect compared with the predominantly quiescent noncultured HSPCs. We previously showed that HSPCs make contact with osteoblasts in vitro via a polarized membrane domain enriched in adhesion molecules such as tetraspanins. Here we show that increased cell cycling during ex vivo culture of HSPCs resulted in disruption of this membrane domain, as evidenced by disruption of polarity of the tetraspanin CD82. Chemical disruption or antibody-mediated blocking of CD82 on noncultured HSPCs resulted in decreased stromal cell adhesion, homing, and engraftment in nonobese diabetic/severe combined immunodeficiency IL-2γ(null) (NSG) mice compared with HSPCs with an intact domain. Most leukemic blasts were actively cycling and correspondingly displayed a loss of domain polarity and decreased homing in NSG mice compared with normal HSPCs. We conclude that quiescent cells, unlike actively cycling cells, display a polarized membrane domain enriched in tetraspanins that mediates homing and engraftment, providing a mechanistic explanation for the homing/engraftment defect of cycling cells and a potential new therapeutic target to enhance engraftment.


Assuntos
Medula Óssea/metabolismo , Transplante de Células-Tronco Hematopoéticas/métodos , Células-Tronco Hematopoéticas/metabolismo , Microdomínios da Membrana/metabolismo , Animais , Antígenos CD34/metabolismo , Ciclo Celular/efeitos dos fármacos , Linhagem Celular Tumoral , Células Cultivadas , Citocinas/farmacologia , Citometria de Fluxo , Imunofluorescência , Humanos , Subunidade gama Comum de Receptores de Interleucina/deficiência , Subunidade gama Comum de Receptores de Interleucina/genética , Proteína Kangai-1/metabolismo , Leucemia/metabolismo , Leucemia/patologia , Camundongos , Camundongos Endogâmicos NOD , Camundongos Knockout , Camundongos SCID , Microscopia Confocal , Receptores CXCR4/metabolismo , Tetraspaninas/metabolismo , Transplante Heterólogo
16.
Commun Integr Biol ; 2(4): 305-7, 2009 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-19721872

RESUMO

Cellular communication within a larger microenvironment is critical for a number of physiological processes. Within the bone marrow niche, direct cell communication between hematopoietic progenitor cells (HPCs) and osteoblasts provides essential cues for their proliferation and survival. While contact-dependent communication between HPCs and osteoblasts is known to be critical, the molecular pathways that govern this interaction are largely unclear. Moreover, the downstream events occurring at the HPC/osteoblast contact site remain uncharacterized, despite their major role in signaling and remodeling within the niche microenvironment. Using live cell imaging approaches, we found that intercellular transfer is a novel mode of cell communication within the bone marrow niche microenvironment. HPCs made prolonged contact with the osteoblast surface via a specialized membrane domain enriched in prominin 1, CD63 and rhodamine PE. At the contact site, portions of the HPC specialized domain containing these molecules were taken up by the osteoblast and internalized into long-lived, SARA-positive, signaling endosomes. This resulted in the down-regulation of Smad signaling by the osteoblasts and a subsequent increase in the production of stromal-derived factor-1 (SDF-1), a chemokine responsible for HPC homing to bone marrow. These findings identify a novel mechanism involving intercellular transfer to signaling endosomes for targeted regulation of signaling and remodeling events within the osteoblastic niche microenvironment.

17.
Nat Cell Biol ; 11(3): 303-11, 2009 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-19198600

RESUMO

Haematopoietic stem-progenitor cells (HSPCs) reside in the bone marrow niche, where interactions with osteoblasts provide essential cues for their proliferation and survival. Here, we use live-cell imaging to characterize both the site of contact between osteoblasts and haematopoietic progenitor cells (HPCs) and events at this site that result in downstream signalling responses important for niche maintenance. HPCs made prolonged contact with the osteoblast surface through a specialized membrane domain enriched in prominin 1, CD63 and rhodamine PE. At the contact site, portions of the specialized domain containing these molecules were taken up by the osteoblast and internalized into SARA-positive signalling endosomes. This caused osteoblasts to downregulate Smad signalling and increase production of stromal-derived factor-1 (SDF-1), a chemokine responsible for HSPC homing to bone marrow. These findings identify a mechanism involving intercellular transfer to signalling endosomes for targeted regulation of signalling and remodelling events within an ex vivo osteoblastic niche.


Assuntos
Medula Óssea/metabolismo , Endossomos/metabolismo , Espaço Extracelular/metabolismo , Transdução de Sinais , Comunicação Celular , Compartimento Celular , Linhagem Celular , Quimiocina CXCL12/biossíntese , Endocitose , Células-Tronco Hematopoéticas/citologia , Células-Tronco Hematopoéticas/metabolismo , Células-Tronco Hematopoéticas/ultraestrutura , Humanos , Microdomínios da Membrana/ultraestrutura , Osteoblastos/citologia , Osteoblastos/metabolismo , Osteoblastos/ultraestrutura , Proteínas Smad/metabolismo
18.
In Vitro Cell Dev Biol Anim ; 44(3-4): 87-95, 2008.
Artigo em Inglês | MEDLINE | ID: mdl-18247100

RESUMO

Osteosarcoma is the most common form of primary bone cancer. In this study, we established a human osteosarcoma cell line (OS 99-1) from a highly aggressive primary tumor. G-banding karyotype analysis demonstrated a large number of clonal abnormalities, as well as extensive intercellular heterogeneity. Through the use of immunologic, molecular, and biochemical analyses, we characterized protein and gene expression profiles confirming the osteogenic nature of the cells. Further evaluation indicated that OS 99-1 cells maintain the capacity to differentiate in an in vitro mineralization assay as well as form tumors in the in vivo chicken embryo model. This cell line provides a useful tool to investigate the molecular mechanisms contributing to osteosarcoma and may have the potential to serve as a culture system for studies involving bone physiology.


Assuntos
Neoplasias Ósseas/patologia , Linhagem Celular Tumoral , Osteossarcoma/patologia , Animais , Calcificação Fisiológica , Diferenciação Celular , Embrião de Galinha , Perfilação da Expressão Gênica , Humanos , Cariotipagem
19.
J Cell Biochem ; 92(4): 820-32, 2004 Jul 01.
Artigo em Inglês | MEDLINE | ID: mdl-15211578

RESUMO

Osteosarcoma is an aggressive primary bone cancer affecting primarily children and young adults. The development of valuable diagnostic indicators and therapeutic agents will be enhanced by the identification and characterization of genes that contribute to its aggressive behavior. We used representational difference analysis to isolate genes differentially expressed between primary human osteosarcoma tumors and subsequent metastatic lung lesions to identify genes potentially involved in metastatic potential. Several genes were differentially expressed between the two tumor populations, including annexin2. The levels of annexin2 mRNA and protein inversely correlated with metastatic potential in a subset of human osteosarcoma tumor specimens, as well as in a human osteosarcoma cell line selected for increased metastatic potential. Annexin2 has been described in several cellular localizations with various functional implications, many of which may be relevant to metastatic potential. Therefore, the subcellular localization of endogenous annexin2 protein was evaluated biochemically by subcellular fractionation and immunologically by flow cytometry and immunofluorescence in osteoblastic cells. Annexin2 was localized to the cytoplasm and intracellular aspect of the plasma membrane, excluded from the nucleus and undetectable on the cell surface or in the conditioned medium. Overexpression of annexin2 in osteosarcoma cells did not alter several in vitro phenotypes often used to assess metastatic potential including motility, adhesion, and proliferation. However, our previous data have implicated annexin2 in the mineralization process of osteoblastic cells in vitro. Consistent with an increase in differentiation-induced mineralization, there was diminished tumorigenicity and experimental metastatic potential of osteosarcoma cells overexpressing annexin2. These data suggest that annexin2 may downregulate osteosarcoma aggressiveness by inducing a more differentiated state in osteoblastic cells.


Assuntos
Anexina A2/metabolismo , Neoplasias Ósseas/metabolismo , Neoplasias Pulmonares/metabolismo , Neoplasias Pulmonares/secundário , Osteossarcoma/metabolismo , Osteossarcoma/secundário , Animais , Anexina A2/genética , Neoplasias Ósseas/patologia , Adesão Celular , Ciclo Celular , Movimento Celular , Regulação para Baixo , Regulação Neoplásica da Expressão Gênica , Humanos , Neoplasias Pulmonares/prevenção & controle , Masculino , Camundongos , Camundongos Nus , RNA Mensageiro/genética , RNA Mensageiro/metabolismo , Transfecção , Células Tumorais Cultivadas
20.
J Cell Sci ; 117(Pt 3): 441-9, 2004 Jan 26.
Artigo em Inglês | MEDLINE | ID: mdl-14679310

RESUMO

While the basic cellular contributions to bone differentiation and mineralization are widely accepted, the regulation of these processes at the intracellular level remains inadequately understood. Our laboratory recently identified annexin 2 as a protein involved in osteoblastic mineralization. Annexin 2 was overexpressed twofold in SaOSLM2 osteoblastic cells as a fusion protein with green fluorescent protein. The overexpression of annexin 2 led to an increase in alkaline phosphatase activity as well as an increase in mineralization. Our data suggest that the increase in alkaline phosphatase activity does not result from increased alkaline phosphatase transcript or protein levels; therefore we evaluated mechanism of action. We determined that both annexin 2 and alkaline phosphatase activity were localized to membrane microdomains called lipid rafts in osteoblastic cells. Annexin 2 overexpression resulted in an increase in alkaline phosphatase activity that was associated with lipid microdomains in a cholesterol-dependent manner. Furthermore, disruption of lipid rafts with a cholesterol sequestering agent or reduction of annexin 2 expression by specific antisense oligonucleotides each resulted in diminished mineralization. Therefore, intact lipid rafts containing annexin 2 appear to be important for alkaline phosphatase activity and may facilitate the osteoblastic mineralization process.


Assuntos
Fosfatase Alcalina/metabolismo , Anexina A2/metabolismo , Calcificação Fisiológica/fisiologia , Colesterol/metabolismo , Microdomínios da Membrana/metabolismo , beta-Ciclodextrinas , Ciclodextrinas/farmacologia , DNA Antissenso , Ativação Enzimática , Humanos , Microscopia de Fluorescência , Osteoblastos/metabolismo , Proteínas Recombinantes de Fusão/metabolismo , Frações Subcelulares , Células Tumorais Cultivadas
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA