Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 20
Filtrar
1.
Proc Natl Acad Sci U S A ; 120(17): e2210735120, 2023 04 25.
Artigo em Inglês | MEDLINE | ID: mdl-37075074

RESUMO

The invasion of nerves by cancer cells, or perineural invasion (PNI), is potentiated by the nerve microenvironment and is associated with adverse clinical outcomes. However, the cancer cell characteristics that enable PNI are poorly defined. Here, we generated cell lines enriched for a rapid neuroinvasive phenotype by serially passaging pancreatic cancer cells in a murine sciatic nerve model of PNI. Cancer cells isolated from the leading edge of nerve invasion showed a progressively increasing nerve invasion velocity with higher passage number. Transcriptome analysis revealed an upregulation of proteins involving the plasma membrane, cell leading edge, and cell movement in the leading neuroinvasive cells. Leading cells progressively became round and blebbed, lost focal adhesions and filipodia, and transitioned from a mesenchymal to amoeboid phenotype. Leading cells acquired an increased ability to migrate through microchannel constrictions and associated more with dorsal root ganglia than nonleading cells. ROCK inhibition reverted leading cells from an amoeboid to mesenchymal phenotype, reduced migration through microchannel constrictions, reduced neurite association, and reduced PNI in a murine sciatic nerve model. Cancer cells with rapid PNI exhibit an amoeboid phenotype, highlighting the plasticity of cancer migration mode in enabling rapid nerve invasion.


Assuntos
Amoeba , Tecido Nervoso , Neoplasias Pancreáticas , Camundongos , Animais , Neoplasias Pancreáticas/genética , Nervo Isquiático/metabolismo , Pâncreas/metabolismo , Tecido Nervoso/metabolismo , Movimento Celular/genética , Invasividade Neoplásica , Microambiente Tumoral
2.
Cell Mol Life Sci ; 74(24): 4405-4420, 2017 12.
Artigo em Inglês | MEDLINE | ID: mdl-28631007

RESUMO

Recent studies have demonstrated a critical role for nerves in enabling tumor progression. The association of nerves with cancer cells is well established for a variety of malignant tumors, including pancreatic, prostate and the head and neck cancers. This association is often correlated with poor prognosis. A strong partnership between cancer cells and nerve cells leads to both cancer progression and expansion of the nerve network. This relationship is supported by molecular pathways related to nerve growth and repair. Peripheral nerves form complex tumor microenvironments, which are made of several cell types including Schwann cells. Recent studies have revealed that Schwann cells enable cancer progression by adopting a de-differentiated phenotype, similar to the Schwann cell response to nerve trauma. A detailed understanding of the molecular and cellular mechanisms involved in the regulation of cancer progression by the nerves is essential to design strategies to inhibit tumor progression.


Assuntos
Neoplasias/patologia , Nervos Periféricos/patologia , Células de Schwann/patologia , Animais , Progressão da Doença , Humanos , Microambiente Tumoral/fisiologia
3.
Proc Natl Acad Sci U S A ; 111(11): 4127-32, 2014 Mar 18.
Artigo em Inglês | MEDLINE | ID: mdl-24591614

RESUMO

Emerging data suggest that in polarized epithelial cells newly synthesized apical and basolateral plasma membrane proteins traffic through different endosomal compartments en route to the respective cell surface. However, direct evidence for trans-endosomal pathways of plasma membrane proteins is still missing and the mechanisms involved are poorly understood. Here, we imaged the entire biosynthetic route of rhodopsin-GFP, an apical marker in epithelial cells, synchronized through recombinant conditional aggregation domains, in live Madin-Darby canine kidney cells using spinning disk confocal microscopy. Our experiments directly demonstrate that rhodopsin-GFP traffics through apical recycling endosomes (AREs) that bear the small GTPase Rab11a before arriving at the apical membrane. Expression of dominant-negative Rab11a drastically reduced apical delivery of rhodopsin-GFP and caused its missorting to the basolateral membrane. Surprisingly, functional inhibition of dynamin-2 trapped rhodopsin-GFP at AREs and caused aberrant accumulation of coated vesicles on AREs, suggesting a previously unrecognized role for dynamin-2 in the scission of apical carrier vesicles from AREs. A second set of experiments, using a unique method to carry out total internal reflection fluorescence microscopy (TIRFM) from the apical side, allowed us to visualize the fusion of rhodopsin-GFP carrier vesicles, which occurred randomly all over the apical plasma membrane. Furthermore, two-color TIRFM showed that Rab11a-mCherry was present in rhodopsin-GFP carrier vesicles and was rapidly released upon fusion onset. Our results provide direct evidence for a role of AREs as a post-Golgi sorting hub in the biosynthetic route of polarized epithelia, with Rab11a regulating cargo sorting at AREs and carrier vesicle docking at the apical membrane.


Assuntos
Vias Biossintéticas/fisiologia , Polaridade Celular/fisiologia , Células Epiteliais/citologia , Rodopsina/metabolismo , Proteínas rab de Ligação ao GTP/metabolismo , Animais , Primers do DNA/genética , Cães , Complexo de Golgi/metabolismo , Immunoblotting , Imuno-Histoquímica , Células Madin Darby de Rim Canino , Microscopia Confocal , Microscopia Eletrônica de Transmissão , Microscopia de Fluorescência , Plasmídeos/genética , Transporte Proteico/fisiologia , Rodopsina/biossíntese , Vesículas Transportadoras/metabolismo
4.
Proc Natl Acad Sci U S A ; 111(19): E2008-17, 2014 May 13.
Artigo em Inglês | MEDLINE | ID: mdl-24778213

RESUMO

The ability of cancer cells to invade along nerves is associated with aggressive disease and diminished patient survival rates. Perineural invasion (PNI) may be mediated by nerve secretion of glial cell line-derived neurotrophic factor (GDNF) attracting cancer cell migration through activation of cell surface Ret proto-oncogene (RET) receptors. GDNF family receptor (GFR)α1 acts as coreceptor with RET, with both required for response to GDNF. We demonstrate that GFRα1 released by nerves enhances PNI, even in the absence of cancer cell GFRα1 expression. Cancer cell migration toward GDNF, RET phosphorylation, and MAPK pathway activity are increased with exposure to soluble GFRα1 in a dose-dependent fashion. Dorsal root ganglia (DRG) release soluble GFRα1, which potentiates RET activation and cancer cell migration. In vitro DRG coculture assays of PNI show diminished PNI with DRG from GFRα1(+/-) mice compared with GFRα1(+/+) mice. An in vivo murine model of PNI demonstrates that cancer cells lacking GFRα1 maintain an ability to invade nerves and impair nerve function, whereas those lacking RET lose this ability. A tissue microarray of human pancreatic ductal adenocarcinomas demonstrates wide variance of cancer cell GFRα1 expression, suggesting an alternate source of GFRα1 in PNI. These findings collectively demonstrate that GFRα1 released by nerves enhances PNI through GDNF-RET signaling and that GFRα1 expression by cancer cells enhances but is not required for PNI. These results advance a mechanistic understanding of PNI and implicate the nerve itself as a key facilitator of this adverse cancer cell behavior.


Assuntos
Adenocarcinoma/metabolismo , Receptores de Fator Neurotrófico Derivado de Linhagem de Célula Glial/metabolismo , Fator Neurotrófico Derivado de Linhagem de Célula Glial/metabolismo , Sistema de Sinalização das MAP Quinases/fisiologia , Neoplasias Pancreáticas/metabolismo , Proteínas Proto-Oncogênicas c-ret/metabolismo , Células 3T3 , Adenocarcinoma/patologia , Animais , Carcinoma Ductal Pancreático/metabolismo , Carcinoma Ductal Pancreático/patologia , Linhagem Celular Tumoral , Movimento Celular/fisiologia , Técnicas de Cocultura , Gânglios Espinais/metabolismo , Gânglios Espinais/patologia , Receptores de Fator Neurotrófico Derivado de Linhagem de Célula Glial/genética , Humanos , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Endogâmicos C57BL , Camundongos Nus , Invasividade Neoplásica , Tecido Nervoso/metabolismo , Tecido Nervoso/patologia , Neoplasias Pancreáticas/patologia , Neoplasias do Sistema Nervoso Periférico/metabolismo , Neoplasias do Sistema Nervoso Periférico/patologia , Proto-Oncogene Mas , RNA Interferente Pequeno/genética , Neuropatia Ciática/metabolismo , Neuropatia Ciática/patologia , Solubilidade
5.
Nature ; 452(7188): 719-23, 2008 Apr 10.
Artigo em Inglês | MEDLINE | ID: mdl-18401403

RESUMO

Clathrin-coated vesicles are vehicles for intracellular trafficking in all nucleated cells, from yeasts to humans. Many studies have demonstrated their essential roles in endocytosis and cellular signalling processes at the plasma membrane. By contrast, very few of their non-endocytic trafficking roles are known, the best characterized being the transport of hydrolases from the Golgi complex to the lysosome. Here we show that clathrin is required for polarity of the basolateral plasma membrane proteins in the epithelial cell line MDCK. Clathrin knockdown depolarized most basolateral proteins, by interfering with their biosynthetic delivery and recycling, but did not affect the polarity of apical proteins. Quantitative live imaging showed that chronic and acute clathrin knockdown selectively slowed down the exit of basolateral proteins from the Golgi complex, and promoted their mis-sorting into apical carrier vesicles. Our results demonstrate a broad requirement for clathrin in basolateral protein trafficking in epithelial cells.


Assuntos
Polaridade Celular , Clatrina/metabolismo , Células Epiteliais/citologia , Animais , Catepsina D/metabolismo , Linhagem Celular , Clatrina/deficiência , Clatrina/genética , Cadeias Pesadas de Clatrina/genética , Cadeias Pesadas de Clatrina/metabolismo , Cães , Células Epiteliais/metabolismo , Complexo de Golgi/metabolismo , Humanos , Inulina/metabolismo , Lisossomos/metabolismo , Transporte Proteico , Receptores de LDL/metabolismo , Receptores da Transferrina/metabolismo , Junções Íntimas/metabolismo , Fatores de Tempo , Rede trans-Golgi/metabolismo
6.
Traffic ; 12(4): 483-98, 2011 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-21199217

RESUMO

Many solute transporters are heterodimers composed of non-glycosylated catalytic and glycosylated accessory subunits. These transporters are specifically polarized to the apical or basolateral membranes of epithelia, but this polarity may vary to fulfill tissue-specific functions. To date, the mechanisms regulating the tissue-specific polarity of heteromeric transporters remain largely unknown. Here, we investigated the sorting signals that determine the polarity of three members of the proton-coupled monocarboxylate transporter (MCT) family, MCT1, MCT3 and MCT4, and their accessory subunit CD147. We show that MCT3 and MCT4 harbor strong redundant basolateral sorting signals (BLSS) in their C-terminal cytoplasmic tails that can direct fusion proteins with the apical marker p75 to the basolateral membrane. In contrast, MCT1 lacks a BLSS and its polarity is dictated by CD147, which contains a weak BLSS that can direct Tac, but not p75 to the basolateral membrane. Knockdown experiments in MDCK cells indicated that basolateral sorting of MCTs was clathrin-dependent but clathrin adaptor AP1B-independent. Our results explain the consistently basolateral localization of MCT3 and MCT4 and the variable localization of MCT1 in different epithelia. They introduce a new paradigm for the sorting of heterodimeric transporters in which a hierarchy of apical and BLSS in the catalytic and/or accessory subunits regulates their tissue-specific polarity.


Assuntos
Polaridade Celular/fisiologia , Epitélio/metabolismo , Transportadores de Ácidos Monocarboxílicos/metabolismo , Sinais Direcionadores de Proteínas/fisiologia , Transporte Proteico/fisiologia , Sequência de Aminoácidos , Animais , Basigina/metabolismo , Membrana Celular/metabolismo , Células Cultivadas , Clatrina/metabolismo , Técnicas de Silenciamento de Genes , Fusão de Membrana/fisiologia , Proteínas de Fusão de Membrana/metabolismo , Proteínas de Membrana Transportadoras/metabolismo , Camundongos , Camundongos Endogâmicos C57BL , Dados de Sequência Molecular , Transportadores de Ácidos Monocarboxílicos/química
7.
Curr Opin Cell Biol ; 17(4): 423-34, 2005 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-15975780

RESUMO

The cell biologist's insight into endosomal diversity, in terms of both form and function, has increased dramatically in the past few years. This understanding has been promoted by the availability of powerful new techniques that allow imaging of both cargo and machinery in the endocytic process in real time, and by our ability to inhibit components of this machinery by RNA interference. The emerging picture from these studies is of a highly complex, dynamic and adaptable endosomal system that interacts at various points with the secretory system of the cell.


Assuntos
Endossomos/classificação , Endossomos/metabolismo , Animais , Citoesqueleto/metabolismo , Lipídeos de Membrana/metabolismo , Transporte Proteico
8.
Adv Biol (Weinh) ; 6(9): e2200089, 2022 09.
Artigo em Inglês | MEDLINE | ID: mdl-35666078

RESUMO

Schwann cells (SCs) are the most abundant cell type in the nerves in the peripheral nervous system and compose a family of subtypes that are endowed with a variety of different functions. SCs facilitate the transmission of neural impulses, provide nutrients and protection for neurons, guide axons in nerve repair, and regulate immune functions. In the context of cancer, recent studies have revealed an active role of SCs in promoting cancer cell invasion, modulating immune responses, and transmitting pain sensation.


Assuntos
Neoplasias , Regeneração Nervosa , Axônios/fisiologia , Neoplasias/metabolismo , Regeneração Nervosa/fisiologia , Sistema Nervoso Periférico , Células de Schwann/metabolismo
9.
J Vis Exp ; (190)2022 12 02.
Artigo em Inglês | MEDLINE | ID: mdl-36533840

RESUMO

Growing evidence suggests that the sympathetic nervous system plays an important role in cancer progression. Adrenergic innervation regulates salivary gland secretion, circadian rhythm, macular degeneration, immune function, and cardiac physiology. Murine surgical sympathectomy is a method for studying the effects of adrenergic innervation by allowing for complete, unilateral adrenergic ablation while avoiding the need for repeated pharmacologic intervention and the associated side effects. However, surgical sympathectomy in mice is technically challenging because of the small size of the superior cervical ganglion. This study describes a surgical technique for reliably identifying and resecting the superior cervical ganglion to ablate the sympathetic nervous system. The successful identification and removal of the ganglion are validated by imaging the fluorescent sympathetic ganglia using a transgenic mouse, identifying post-resection Horner's syndrome, staining for adrenergic markers in the resected ganglia, and observing diminished adrenergic immunofluorescence in the target organs following sympathectomy. This model enables future studies of cancer progression as well as other physiological processes regulated by the sympathetic nervous system.


Assuntos
Ganglionectomia , Simpatectomia , Animais , Camundongos , Modelos Animais de Doenças , Gânglios Simpáticos/cirurgia , Sistema Nervoso Simpático/cirurgia , Sistema Nervoso Simpático/fisiologia , Gânglio Cervical Superior/cirurgia , Adrenérgicos
10.
Cancer Discov ; 12(10): 2454-2473, 2022 10 05.
Artigo em Inglês | MEDLINE | ID: mdl-35881881

RESUMO

Nerves are a component of the tumor microenvironment contributing to cancer progression, but the role of cells from nerves in facilitating cancer invasion remains poorly understood. Here we show that Schwann cells (SC) activated by cancer cells collectively function as tumor-activated Schwann cell tracks (TAST) that promote cancer cell migration and invasion. Nonmyelinating SCs form TASTs and have cell gene expression signatures that correlate with diminished survival in patients with pancreatic ductal adenocarcinoma. In TASTs, dynamic SCs form tracks that serve as cancer pathways and apply forces on cancer cells to enhance cancer motility. These SCs are activated by c-Jun, analogous to their reprogramming during nerve repair. This study reveals a mechanism of cancer cell invasion that co-opts a wound repair process and exploits the ability of SCs to collectively organize into tracks. These findings establish a novel paradigm of how cancer cells spread and reveal therapeutic opportunities. SIGNIFICANCE: How the tumor microenvironment participates in pancreatic cancer progression is not fully understood. Here, we show that SCs are activated by cancer cells and collectively organize into tracks that dynamically enable cancer invasion in a c-Jun-dependent manner. See related commentary by Amit and Maitra, p. 2240. This article is highlighted in the In This Issue feature, p. 2221.


Assuntos
Carcinoma Ductal Pancreático , Neoplasias Pancreáticas , Carcinoma Ductal Pancreático/patologia , Movimento Celular/genética , Humanos , Neoplasias Pancreáticas/patologia , Células de Schwann/metabolismo , Microambiente Tumoral , Neoplasias Pancreáticas
11.
Nat Cancer ; 1: 1027-1031, 2021 11.
Artigo em Inglês | MEDLINE | ID: mdl-34327335

RESUMO

Recent advances in cancer neuroscience necessitate the systematic analysis of neural influences in cancer as potential therapeutic targets in oncology. Here, we outline recommendations for future preclinical and translational research in this field.


Assuntos
Neoplasias , Neurociências , Previsões , Humanos , Neoplasias/terapia , Pesquisa Translacional Biomédica
12.
Mol Cancer Res ; 18(6): 913-925, 2020 06.
Artigo em Inglês | MEDLINE | ID: mdl-32086369

RESUMO

Perineural invasion (PNI) is an ominous form of cancer progression along nerves associated with poor clinical outcome. Glial derived neurotrophic factor (GDNF) interacts with cancer cell RET receptors to enable PNI, but downstream events remain undefined. We demonstrate that GDNF leads to early activation of the GTPase Cdc42 in pancreatic cancer cells, but only delayed activation of RhoA and does not affect Rac1. Depletion of Cdc42 impairs pancreatic cancer cell chemotaxis toward GDNF and nerves. An siRNA library of guanine nucleotide exchange factors was screened to identify activators of Cdc42. ARHGEF7 (ß-Pix) was required for Cdc42 activation and chemotaxis toward nerves, and also colocalizes with RET under GDNF stimulation. Cdc42 enables PNI in an in vitro dorsal root ganglia coculture model, and controls the directionality of migration but does not affect cell speed or cell viability. In contrast, Rac1 was necessary for cell speed but not directionality, while the RhoA was not necessary for either cell speed or directionality. Cdc42 was required for PNI in an in vivo murine sciatic nerve model. Depletion of Cdc42 significantly diminished the length of PNI, volume of PNI, and motor nerve paralysis resulting from PNI. Activated Cdc42 is expressed in human salivary ductal cancer cells invading nerves. These findings establish the GDNF-RET-ß-Pix-Cdc42 pathway as a directional regulator of pancreatic cancer cell migration toward nerves, highlight the importance of directional migration in PNI, and offer novel targets for therapy. IMPLICATIONS: Cdc42 regulates cancer cell directional migration toward and along nerves in PNI.


Assuntos
Movimento Celular , Fator Neurotrófico Derivado de Linhagem de Célula Glial/metabolismo , Neoplasias Pancreáticas/patologia , Fatores de Troca de Nucleotídeo Guanina Rho/metabolismo , Nervo Isquiático/patologia , Proteína cdc42 de Ligação ao GTP/metabolismo , Animais , Apoptose , Proliferação de Células , Feminino , Fator Neurotrófico Derivado de Linhagem de Célula Glial/genética , Humanos , Camundongos , Camundongos Nus , Invasividade Neoplásica , Neoplasias Pancreáticas/genética , Neoplasias Pancreáticas/metabolismo , Fatores de Troca de Nucleotídeo Guanina Rho/genética , Nervo Isquiático/metabolismo , Células Tumorais Cultivadas , Ensaios Antitumorais Modelo de Xenoenxerto , Proteína cdc42 de Ligação ao GTP/genética
13.
Cancer Cell ; 38(1): 11-14, 2020 07 13.
Artigo em Inglês | MEDLINE | ID: mdl-32531270

RESUMO

Neuro-glial activation is a recently identified hallmark of growing cancers. Targeting tumor hyperinnervation in preclinical and small clinical trials has yielded promising antitumor effects, highlighting the need of systematic analysis of neural influences in cancer (NIC). Here, we outline the strategies translating these findings from bench to the clinic.


Assuntos
Neoplasias/fisiopatologia , Neoplasias/terapia , Sistema Nervoso/fisiopatologia , Dor do Câncer/diagnóstico , Dor do Câncer/fisiopatologia , Dor do Câncer/terapia , Denervação/métodos , Humanos , Neoplasias/diagnóstico
14.
J Vis Exp ; (134)2018 04 23.
Artigo em Inglês | MEDLINE | ID: mdl-29733315

RESUMO

Cancer cells invade nerves through a process termed perineural invasion (PNI), in which cancer cells proliferate and migrate in the nerve microenvironment. This type of invasion is exhibited by a variety of cancer types, and very frequently is found in pancreatic cancer. The microscopic size of nerve fibers within mouse pancreas renders the study of PNI difficult in orthotopic murine models. Here, we describe a heterotopic in vivo model of PNI, where we inject syngeneic pancreatic cancer cell line Panc02-H7 into the murine sciatic nerve. In this model, sciatic nerves of anesthetized mice are exposed and injected with cancer cells. The cancer cells invade in the nerves proximally toward the spinal cord from the point of injection. The invaded sciatic nerves are then extracted and processed with OCT for frozen sectioning. H&E and immunofluorescence staining of these sections allow quantification of both the degree of invasion and changes in protein expression. This model can be applied to a variety of studies on PNI given its versatility. Using mice with different genetic modifications and/or different types of cancer cells allows for investigation of the cellular and molecular mechanisms of PNI and for different cancer types. Furthermore, the effects of therapeutic agents on nerve invasion can be studied by applying treatment to these mice.


Assuntos
Adenocarcinoma/genética , Adenocarcinoma/metabolismo , Tecido Nervoso/crescimento & desenvolvimento , Neoplasias Pancreáticas/genética , Neoplasias Pancreáticas/metabolismo , Nervos Periféricos/crescimento & desenvolvimento , Nervo Isquiático/patologia , Animais , Feminino , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Invasividade Neoplásica
15.
Cancer Res ; 77(22): 6400-6414, 2017 11 15.
Artigo em Inglês | MEDLINE | ID: mdl-28951461

RESUMO

Perineural invasion (PNI) is an ominous event strongly linked to poor clinical outcome. Cells residing within peripheral nerves collaborate with cancer cells to enable PNI, but the contributing conditions within the tumor microenvironment are not well understood. Here, we show that CCR2-expressing inflammatory monocytes (IM) are preferentially recruited to sites of PNI, where they differentiate into macrophages and potentiate nerve invasion through a cathepsin B-mediated process. A series of adoptive transfer experiments with genetically engineered donors and recipients demonstrated that IM recruitment to nerves was driven by CCL2 released from Schwann cells at the site of PNI, but not CCL7, an alternate ligand for CCR2. Interruption of either CCL2-CCR2 signaling or cathepsin B function significantly impaired PNI in vivo Correlative studies in human specimens demonstrated that cathepsin B-producing macrophages were enriched in invaded nerves, which was associated with increased local tumor recurrence. These findings deepen our understanding of PNI pathogenesis and illuminate how PNI is driven in part by corruption of a nerve repair program. Further, they support the exploration of inhibiting IM recruitment and function as a targeted therapy for PNI. Cancer Res; 77(22); 6400-14. ©2017 AACR.


Assuntos
Catepsina B/metabolismo , Quimiocina CCL2/metabolismo , Monócitos/metabolismo , Neoplasias Pancreáticas/metabolismo , Nervos Periféricos/metabolismo , Animais , Linhagem Celular , Linhagem Celular Tumoral , Quimiocina CCL2/genética , Humanos , Macrófagos/metabolismo , Camundongos Endogâmicos C57BL , Camundongos Knockout , Camundongos Nus , Monócitos/patologia , Invasividade Neoplásica , Neoplasias Experimentais/genética , Neoplasias Experimentais/metabolismo , Neoplasias Experimentais/patologia , Neoplasias Pancreáticas/genética , Neoplasias Pancreáticas/patologia , Nervos Periféricos/patologia , Receptores CCR2/genética , Receptores CCR2/metabolismo , Células de Schwann/metabolismo , Transplante Heterólogo
16.
J Clin Invest ; 126(4): 1538-54, 2016 Apr 01.
Artigo em Inglês | MEDLINE | ID: mdl-26999607

RESUMO

Nerves enable cancer progression, as cancers have been shown to extend along nerves through the process of perineural invasion, which carries a poor prognosis. Furthermore, the innervation of some cancers promotes growth and metastases. It remains unclear, however, how nerves mechanistically contribute to cancer progression. Here, we demonstrated that Schwann cells promote cancer invasion through direct cancer cell contact. Histological evaluation of murine and human cancer specimens with perineural invasion uncovered a subpopulation of Schwann cells that associates with cancer cells. Coculture of cancer cells with dorsal root ganglion extracts revealed that Schwann cells direct cancer cells to migrate toward nerves and promote invasion in a contact-dependent manner. Upon contact, Schwann cells induced the formation of cancer cell protrusions in their direction and intercalated between the cancer cells, leading to cancer cell dispersion. The formation of these processes was dependent on Schwann cell expression of neural cell adhesion molecule 1 (NCAM1) and ultimately promoted perineural invasion. Moreover, NCAM1-deficient mice showed decreased neural invasion and less paralysis. Such Schwann cell behavior reflects normal Schwann cell programs that are typically activated in nerve repair but are instead exploited by cancer cells to promote perineural invasion and cancer progression.


Assuntos
Antígeno CD56/metabolismo , Neoplasias Experimentais/metabolismo , Células de Schwann/metabolismo , Animais , Linhagem Celular Tumoral , Técnicas de Cocultura , Humanos , Camundongos , Camundongos Nus , Células NIH 3T3 , Invasividade Neoplásica , Neoplasias Experimentais/patologia , Células de Schwann/patologia
17.
Mol Cancer Res ; 13(2): 380-90, 2015 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-25312961

RESUMO

UNLABELLED: Perineural invasion is a form of cancer progression where cancer cells invade along nerves. This behavior is associated with poor clinical outcomes; therefore, it is critical to identify novel ligand-receptor interactions between nerves and cancer cells that support the process of perineural invasion. A proteomic profiler chemokine array was used to screen for nerve-derived factors secreted from tissue explants of dorsal root ganglion (DRG), and CCL2 was identified as a lead candidate. Prostate cancer cell line expression of CCR2, the receptor to CCL2, correlated closely with MAPK and Akt pathway activity and cell migration towards CCL2 and DRG. In vitro nerve and cancer coculture invasion assays of perineural invasion demonstrated that cancer cell CCR2 expression facilitates perineural invasion. Perineural invasion is significantly diminished in coculture assays when using DRG harvested from CCL2(-/-) knockout mice as compared with control CCL2(+/+) mice, indicating that CCR2 is required for perineural invasion in this murine model of perineural invasion. Furthermore, 20 of 21 (95%) patient specimens of prostate adenocarcinoma with perineural invasion exhibited CCR2 expression by immunohistochemistry, while just 3 of 13 (23%) lacking perineural invasion expressed CCR2. In summary, nerve-released CCL2 supports prostate cancer migration and perineural invasion though CCR2-mediated signaling. IMPLICATIONS: These results reveal CCL2-CCR2 signaling as a key ligand-receptor mechanism that mediates cancer cell communication with nerves during perineural invasion and highlight a potential future therapeutic target.


Assuntos
Quimiocina CCL2/metabolismo , Gânglios Espinais/patologia , Neoplasias da Próstata/patologia , Receptores CCR2/metabolismo , Animais , Linhagem Celular Tumoral , Células Cultivadas , Técnicas de Cocultura , Gânglios Espinais/citologia , Gânglios Espinais/metabolismo , Humanos , Masculino , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Endogâmicos C57BL , Invasividade Neoplásica , Neoplasias da Próstata/metabolismo , Transdução de Sinais
18.
Dev Cell ; 22(4): 811-23, 2012 Apr 17.
Artigo em Inglês | MEDLINE | ID: mdl-22516199

RESUMO

Clathrin and the epithelial-specific clathrin adaptor AP-1B mediate basolateral trafficking in epithelia. However, several epithelia lack AP-1B, and mice knocked out for AP-1B are viable, suggesting the existence of additional mechanisms that control basolateral polarity. Here, we demonstrate a distinct role of the ubiquitous clathrin adaptor AP-1A in basolateral protein sorting. Knockdown of AP-1A causes missorting of basolateral proteins in MDCK cells, but only after knockdown of AP-1B, suggesting that AP-1B can compensate for lack of AP-1A. AP-1A localizes predominantly to the TGN, and its knockdown promotes spillover of basolateral proteins into common recycling endosomes, the site of function of AP-1B, suggesting complementary roles of both adaptors in basolateral sorting. Yeast two-hybrid assays detect interactions between the basolateral signal of transferrin receptor and the medium subunits of both AP-1A and AP-1B. The basolateral sorting function of AP-1A reported here establishes AP-1 as a major regulator of epithelial polarity.


Assuntos
Complexo 1 de Proteínas Adaptadoras/metabolismo , Polaridade Celular , Clatrina/metabolismo , Endossomos/metabolismo , Células Epiteliais/metabolismo , Rede trans-Golgi/fisiologia , Complexo 1 de Proteínas Adaptadoras/antagonistas & inibidores , Complexo 1 de Proteínas Adaptadoras/genética , Animais , Membrana Celular/metabolismo , Células Cultivadas , Cães , Imunofluorescência , Transporte Proteico , RNA Interferente Pequeno/genética , Receptores de LDL/metabolismo , Receptores da Transferrina/metabolismo , Técnicas do Sistema de Duplo-Híbrido
19.
Mol Biol Cell ; 20(1): 438-51, 2009 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-18987335

RESUMO

The functions of the actin cytoskeleton in post-Golgi trafficking are still poorly understood. Here, we report the role of LIM Kinase 1 (LIMK1) and its substrate cofilin in the trafficking of apical and basolateral proteins in Madin-Darby canine kidney cells. Our data indicate that LIMK1 and cofilin organize a specialized population of actin filaments at the Golgi complex that is selectively required for the emergence of an apical cargo route to the plasma membrane (PM). Quantitative pulse-chase live imaging experiments showed that overexpression of kinase-dead LIMK1 (LIMK1-KD), or of LIMK1 small interfering RNA, or of an activated cofilin mutant (cofilin S3A), selectively slowed down the exit from the trans-Golgi network (TGN) of the apical PM marker p75-green fluorescent protein (GFP) but did not interfere with the apical PM marker glycosyl phosphatidylinositol-YFP or the basolateral PM marker neural cell adhesion molecule-GFP. High-resolution live imaging experiments of carrier formation and release by the TGN and analysis of peri-Golgi actin dynamics using photoactivatable GFP suggest a scenario in which TGN-localized LIMK1-cofilin regulate a population of actin filaments required for dynamin-syndapin-cortactin-dependent generation and/or fission of precursors to p75 transporters.


Assuntos
Fatores de Despolimerização de Actina/metabolismo , Dinaminas/metabolismo , Quinases Lim/metabolismo , Rede trans-Golgi/metabolismo , Fatores de Despolimerização de Actina/genética , Actinas/metabolismo , Animais , Biomarcadores/metabolismo , Linhagem Celular , Polaridade Celular , Citoesqueleto/metabolismo , Cães , Dinaminas/genética , Complexo de Golgi/metabolismo , Isoenzimas/genética , Isoenzimas/metabolismo , Quinases Lim/genética , Modelos Biológicos , Moléculas de Adesão de Célula Nervosa/genética , Moléculas de Adesão de Célula Nervosa/metabolismo , Transporte Proteico/fisiologia , RNA Interferente Pequeno/genética , RNA Interferente Pequeno/metabolismo , Receptor de Fator de Crescimento Neural/genética , Receptor de Fator de Crescimento Neural/metabolismo , Proteínas Recombinantes de Fusão/genética , Proteínas Recombinantes de Fusão/metabolismo
20.
Endocrine ; 19(1): 13-22, 2002 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-12583599

RESUMO

One of the primary regulators of maternofetal glucose transfer is the density of glucose transporter proteins in the placenta. These transporters, members of the GLUT gene family of facilitated-diffusion transporters, are embedded in the microvillous (maternal-facing) and basal (fetal-facing) membranes of the syncytiotrophoblast, the main placental barrier layer. Eight members of this family have been described in human placental tissue, but only GLUT1 protein has been identified in the syncytium, where its distribution is asymmetric. The microvillous membrane contains markedly more transporter than the basal, and, as a result, the basal membrane acts as the rate-limiting step in transplacental glucose transport; thus, changes in the density of basal membrane GLUT1 will have a significant impact on transplacental glucoseflux. What little is known about syncytial GLUT1 expression is restricted to factors associated with fetoplacental growth and metabolism; GLUT is inversely regulated by glucose concentration and basal membrane GLUT1 is positively regulated by insulin-like growth factor I, placental growth hormone, and hypoxia. In vivo, basal membrane GLUT1 is upregulated over gestation, increased in diabetic pregnancy, and decreased in chronic hypoxia, while microvillous membrane GLUT1 is unaffected. The contrast between in vitro and in vivo regulation and the specific changes in GLUT1 distribution suggest more complex regulatory interactions than those yet described.


Assuntos
Desenvolvimento Embrionário e Fetal , Glucose/metabolismo , Proteínas Musculares , Proteínas do Tecido Nervoso , Placenta/metabolismo , Transporte Biológico , Feminino , Transportador de Glucose Tipo 1 , Transportador de Glucose Tipo 3 , Transportador de Glucose Tipo 4 , Humanos , Proteínas de Transporte de Monossacarídeos/metabolismo , Gravidez
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA