Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 11 de 11
Filtrar
Mais filtros

Base de dados
Tipo de documento
Intervalo de ano de publicação
1.
Biochim Biophys Acta Mol Cell Res ; 1868(11): 119101, 2021 10.
Artigo em Inglês | MEDLINE | ID: mdl-34280426

RESUMO

A critical unknown in the field of skeletal metastases is how cancer cells find a way to thrive under harsh conditions, as exemplified by metastatic colonization of adipocyte-rich bone marrow by prostate carcinoma cells. To begin understanding molecular processes that enable tumor cells to survive and progress in difficult microenvironments such as bone, we performed unbiased examination of the transcriptome of two different prostate cancer cell lines in the absence or presence of bone marrow adipocytes. Our RNAseq analyses and subsequent quantitative PCR and protein-based assays reveal that upregulation of endoplasmic reticulum (ER) stress and unfolded protein response (UPR) genes is a shared signature between metastatic prostate carcinoma cell lines of different origin. Pathway analyses and pharmacological examinations highlight the ER chaperone BIP as an upstream coordinator of this transcriptomic signature. Additional patient-based data support our overall conclusion that ER stress and UPR induction are shared, important factors in the response and adaptation of metastatic tumor cells to their micro-environment. Our studies pave the way for additional mechanistic investigations and offer new clues towards effective therapeutic interventions in metastatic disease.


Assuntos
Adipócitos/metabolismo , Neoplasias da Próstata/genética , Animais , Células Cultivadas , Chaperona BiP do Retículo Endoplasmático , Estresse do Retículo Endoplasmático/genética , Humanos , Masculino , Camundongos , Neoplasias da Próstata/metabolismo , Neoplasias da Próstata/patologia , RNA-Seq , Transcriptoma , Resposta a Proteínas não Dobradas/genética
2.
ACS Chem Biol ; 14(12): 2833-2840, 2019 12 20.
Artigo em Inglês | MEDLINE | ID: mdl-31750642

RESUMO

Acquired resistance to apoptotic agents is a long-standing challenge in cancer treatment. Cathepsin B (CTSB) is an enzyme which, among many essential functions, promotes apoptosis during cellular stress through regulation of intracellular proteolytic networks on the minute time scale. Recent data indicate that CTSB inhibition may be a promising method to steer cells away from apoptotic death toward necrosis, a mechanism of cell death that can overcome resistance to apoptotic agents, stimulate an immune response and promote antitumor immunity. Unfortunately, rapid and selective intracellular inactivation of CTSB has not been possible. However, here we report on the synthesis and characterization of photochemical and biological properties of BODIPY-caged inhibitors of CTSB that are cell permeable, highly selective and activated rapidly upon exposure to visible light. Intriguingly, these compounds display tunable photophysical and biological properties based on substituents bound directly to boron. Me2BODIPY-caged compound 8 displays the dual-action capability of light-accelerated CTSB inhibition and singlet oxygen production from a singular molecular entity. The dual-action capacity of 8 leads to a rapid necrotic response in MDA-MB-231 triple negative breast cancer cells with high phototherapeutic indexes (>30) and selectivity vs noncancerous cells that neither CTSB inhibition nor photosensitization gives alone. Our work confirms that singlet oxygen production and CTSB inactivation is highly synergistic and a promising method for killing cancer cells. Furthermore, this ability to trigger intracellular inactivation of CTSB with light provides researchers with a powerful photochemical tool for probing biochemical processes on short time scales.


Assuntos
Apoptose/efeitos dos fármacos , Compostos de Boro/química , Catepsina B/antagonistas & inibidores , Inibidores de Cisteína Proteinase/farmacologia , Luz , Neoplasias/patologia , Linhagem Celular Tumoral , Inibidores de Cisteína Proteinase/química , Humanos , Estresse Oxidativo
3.
Mol Cancer Res ; 17(12): 2508-2521, 2019 12.
Artigo em Inglês | MEDLINE | ID: mdl-31562254

RESUMO

Adipocyte-tumor cell cross-talk is one of the critical mediators of tumor progression and an emerging facilitator of therapy evasion. Tumor cells that metastasize to adipocyte-rich bone marrow take advantage of the interplay between metabolic and inflammatory pathways to activate prosurvival mechanisms that allow them to thrive and escape therapy. Using in vitro and in vivo models of marrow adiposity, we demonstrate that metastatic prostate carcinoma cells engage bone marrow adipocytes in a functional cross-talk that promotes IL1ß expression in tumor cells. Tumor-supplied IL1ß contributes to adipocyte lipolysis and regulates a proinflammatory phenotype in adipocytes via upregulation of COX-2 and MCP-1. We further show that the enhanced activity of the IL1ß/COX-2/MCP-1 axis and a resulting increase in PGE2 production by adipocytes coincide with augmented hypoxia signaling and activation of prosurvival pathways in tumor cells, revealing a potential mechanism of chemoresistance. The major consequence of this interplay is the reduced response of prostate cancer cells to docetaxel, a phenomenon sensitive to the inhibition of lipolysis. IMPLICATIONS: Studies presented herein highlight adipocyte lipolysis as a tumor-regulated metabolic event that engages proinflammatory cross-talk in the microenvironment to promote prostate cancer progression in bone. Understanding the impact of bone marrow adipose tissue on tumor adaptation, survival, and chemotherapy response is fundamentally important, as current treatment options for metastatic prostate cancer are palliative.


Assuntos
Quimiocina CCL2/genética , Ciclo-Oxigenase 2/genética , Interleucina-1beta/genética , Neoplasias da Próstata/tratamento farmacológico , Adipócitos/efeitos dos fármacos , Animais , Biópsia , Células da Medula Óssea/efeitos dos fármacos , Dinoprostona/genética , Docetaxel/farmacologia , Resistencia a Medicamentos Antineoplásicos/efeitos dos fármacos , Humanos , Inflamação/tratamento farmacológico , Inflamação/genética , Inflamação/patologia , Lipólise/efeitos dos fármacos , Masculino , Camundongos , Próstata/efeitos dos fármacos , Próstata/patologia , Neoplasias da Próstata/genética , Neoplasias da Próstata/patologia , Transdução de Sinais/efeitos dos fármacos , Microambiente Tumoral/efeitos dos fármacos
4.
Curr Osteoporos Rep ; 16(4): 443-457, 2018 08.
Artigo em Inglês | MEDLINE | ID: mdl-29869753

RESUMO

PURPOSE OF REVIEW: Bone marrow adipocytes have emerged in recent years as key contributors to metastatic progression in bone. In this review, we focus specifically on their role as the suppliers of lipids and discuss pro-survival pathways that are closely linked to lipid metabolism, affected by the adipocyte-tumor cell interactions, and likely impacting the ability of the tumor cell to thrive in bone marrow space and evade therapy. RECENT FINDINGS: The combined in silico, pre-clinical, and clinical evidence shows that in adipocyte-rich tissues such as bone marrow, tumor cells rely on exogenous lipids for regulation of cellular energetics and adaptation to harsh metabolic conditions of the metastatic niche. Adipocyte-supplied lipids have a potential to alter the cell's metabolic decisions by regulating glycolysis and respiration, fatty acid oxidation, lipid desaturation, and PPAR signaling. The downstream effects of lipid signaling on mitochondrial homeostasis ultimately control life vs. death decisions, providing a mechanism for gaining survival advantage and reduced sensitivity to treatment. There is a need for future research directed towards identifying the key metabolic and signaling pathways that regulate tumor dependence on exogenous lipids and consequently drive the pro-survival behavior in the bone marrow niche.


Assuntos
Adipócitos/metabolismo , Células da Medula Óssea/metabolismo , Neoplasias Ósseas/metabolismo , Metabolismo dos Lipídeos , Microambiente Tumoral , Adipócitos/citologia , Apoptose , Células da Medula Óssea/citologia , Neoplasias Ósseas/secundário , Respiração Celular , Ácidos Graxos/metabolismo , Glicerol/metabolismo , Glicólise , Humanos , Mitocôndrias/metabolismo , Oxirredução , Fosforilação Oxidativa , Estresse Oxidativo , Receptores Ativados por Proliferador de Peroxissomo/metabolismo , Transdução de Sinais , Hipóxia Tumoral
5.
Sci Rep ; 8(1): 40, 2018 01 08.
Artigo em Inglês | MEDLINE | ID: mdl-29311669

RESUMO

Metastatic tumor cells engage the local tumor microenvironment and activate specific pro-survival mechanisms to thrive and progress in the harsh bone marrow niche. Here we show that the major contributors to the survival of carcinoma cells that have colonized the bone marrow are the adipocyte-induced oxidative stress and ER stress pathways. We demonstrate that upon exposure to adipocyte-rich environments in vitro or in vivo, bone-trophic prostate and breast tumor cells upregulate the oxidative stress enzyme, HO-1. We also show that HO-1 levels are significantly increased in human metastatic prostate cancer tissues and that stable HO-1 overexpression in tumor cells promotes growth and invasiveness. Co-incident with the adipocyte-induced expression of HO-1, there is an upregulation of ER chaperone BIP and splicing of XBP1, indicating adipocyte-driven unfolded protein response, a process that we show to be sensitive to antioxidant treatment. Importantly, we also demonstrate that triggering of the oxidative stress and ER stress responses, or HO-1 induction by adipocyte exposure result in the activation of pro-survival pathways, involving survivin. Collectively, our findings reveal a new link between HO-1 and survivin expression in tumor cells, and provide a new insight into potentially targetable survival pathways in bone-metastatic disease.


Assuntos
Adipócitos/metabolismo , Osso e Ossos/metabolismo , Estresse do Retículo Endoplasmático , Regulação da Expressão Gênica , Heme Oxigenase-1/genética , Proteínas Inibidoras de Apoptose/genética , Estresse Oxidativo , Animais , Biópsia , Neoplasias Ósseas/genética , Neoplasias Ósseas/metabolismo , Neoplasias Ósseas/patologia , Linhagem Celular Tumoral , Sobrevivência Celular/genética , Heme Oxigenase-1/metabolismo , Humanos , Imuno-Histoquímica , Proteínas Inibidoras de Apoptose/metabolismo , Masculino , Camundongos , Oligopeptídeos/metabolismo , Survivina , Proteína 1 de Ligação a X-Box/metabolismo
6.
Oncotarget ; 7(40): 64854-64877, 2016 10 04.
Artigo em Inglês | MEDLINE | ID: mdl-27588494

RESUMO

Metabolic adaptation is increasingly recognized as a key factor in tumor progression, yet its involvement in metastatic bone disease is not understood. Bone is as an adipocyte-rich organ, and a major site of metastasis from prostate cancer. Bone marrow adipocytes are metabolically active cells capable of shaping tumor metabolism via lipolysis and lipid transfer. In this study, using in vitro and in vivo models of marrow adiposity, we demonstrate that marrow fat cells promote Warburg phenotype in metastatic prostate cancer cells. We show increased expression of glycolytic enzymes, increased lactate production, and decreased mitochondrial oxidative phosphorylation in tumor cells exposed to adipocytes that require paracrine signaling between the two cell types. We also reveal that prostate cancer cells are capable of inducing adipocyte lipolysis as a postulated mechanism of sustenance. We provide evidence that adipocytes drive metabolic reprogramming of tumor cells via oxygen-independent mechanism of HIF-1α activation that can be reversed by HIF-1α downregulation. Importantly, we also demonstrate that the observed metabolic signature in tumor cells exposed to adipocytes mimics the expression patterns seen in patients with metastatic disease. Together, our data provide evidence for a functional relationship between marrow adipocytes and tumor cells in bone that has likely implications for tumor growth and survival within the metastatic niche.


Assuntos
Adipócitos/patologia , Células da Medula Óssea/patologia , Neoplasias Ósseas/metabolismo , Carcinogênese/genética , Subunidade alfa do Fator 1 Induzível por Hipóxia/metabolismo , Neoplasias da Próstata/metabolismo , Síndrome de Walker-Warburg/genética , Adipócitos/metabolismo , Animais , Células da Medula Óssea/metabolismo , Neoplasias Ósseas/genética , Neoplasias Ósseas/secundário , Técnicas de Cocultura , Glicólise/genética , Humanos , Subunidade alfa do Fator 1 Induzível por Hipóxia/genética , Metabolismo dos Lipídeos , Lipólise/genética , Masculino , Camundongos , Comunicação Parácrina , Neoplasias da Próstata/genética , Neoplasias da Próstata/patologia , RNA Interferente Pequeno/genética , Transcriptoma , Células Tumorais Cultivadas , Ensaios Antitumorais Modelo de Xenoenxerto
7.
Biol Chem ; 397(6): 571-82, 2016 06 01.
Artigo em Inglês | MEDLINE | ID: mdl-26901495

RESUMO

Collagenolytic activity of cathepsin K is important for many physiological and pathological processes including osteoclast-mediated bone degradation, macrophage function and fibroblast-mediated matrix remodeling. Here, we report application of a light-activated inhibitor for controlling activity of cathepsin K in a 3D functional imaging assay. Using prostate carcinoma cell line engineered to overexpress cathepsin K, we demonstrate the utility of the proteolytic assay in living tumor spheroids for the evaluation and quantification of the inhibitor effects on cathepsin K-mediated collagen I degradation. Importantly, we also show that utilizing the ruthenium-caged version of a potent nitrile cathepsin K inhibitor (4), cis-[Ru(bpy)2(4)2](BF4)2 (5), offers significant advantage in terms of effective concentration of the inhibitor and especially its light-activated control in the 3D assay. Our results suggest that light activation provides a suitable, attractive approach for spatial and temporal control of proteolytic activity, which remains a critical, unmet need in treatment of human diseases, especially cancer.


Assuntos
Catepsina K/antagonistas & inibidores , Imageamento Tridimensional , Luz , Neoplasias da Próstata/patologia , Inibidores de Proteases/farmacologia , Catepsina K/genética , Linhagem Celular Tumoral , Colágeno Tipo I/metabolismo , Regulação Neoplásica da Expressão Gênica/efeitos dos fármacos , Regulação Neoplásica da Expressão Gênica/efeitos da radiação , Humanos , Masculino , Neoplasias da Próstata/genética , Proteólise/efeitos dos fármacos , Proteólise/efeitos da radiação
8.
Clin Exp Metastasis ; 32(4): 353-68, 2015 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-25802102

RESUMO

Increased bone marrow adiposity is a common feature of advanced age, obesity and associated metabolic pathologies. Augmented numbers of marrow adipocytes positively correlate with dysregulated bone remodeling, also a well-established complication of metastatic disease. We have shown previously that marrow adiposity accelerates prostate tumor progression in the skeleton and promotes extensive destruction of the bone; however, the factors behind adipocyte-driven osteolysis in the skeletal tumor microenvironment are not currently known. In this study, utilizing in vivo diet-induced models of bone marrow adiposity, we reveal evidence for positive correlation between increased marrow fat content, bone degradation by ARCaP(M) and PC3 prostate tumors, and augmented levels of host-derived CXCL1 and CXCL2, ligands of CXCR2 receptor. We show by in vitro osteoclastogenesis assays that media conditioned by bone marrow adipocytes is a significant source of CXCL1 and CXCL2 proteins. We also demonstrate that both the adipocyte-conditioned media and the recombinant CXCL1 and CXCL2 ligands efficiently accelerate osteoclast maturation, a process that can be blocked by neutralizing antibodies to each of the chemokines. We further confirm the contribution of CXCR2 signaling axis to adiposity-driven osteoclastogenesis by blocking fat cell-induced osteoclast differentiation with CXCR2 antagonist or neutralizing antibodies. Together, our results link CXCL1 and CXCL2 chemokines with bone marrow adiposity and implicate CXCR2 signaling in promoting effects of marrow fat on progression of skeletal tumors in bone.


Assuntos
Neoplasias Ósseas/secundário , Quimiocina CXCL1/metabolismo , Quimiocina CXCL2/metabolismo , Osteólise/patologia , Neoplasias da Próstata/patologia , Adipócitos/metabolismo , Adiposidade , Animais , Medula Óssea/metabolismo , Células da Medula Óssea/metabolismo , Remodelação Óssea/fisiologia , Osso e Ossos/patologia , Catepsina K/metabolismo , Diferenciação Celular , Linhagem Celular Tumoral , Técnicas de Cocultura , Humanos , Masculino , Camundongos , Transplante de Neoplasias , Obesidade , Osteoclastos/citologia , Osteogênese/fisiologia , Transplante Heterólogo , Microambiente Tumoral
9.
Cancer Metastasis Rev ; 33(2-3): 527-43, 2014 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-24398857

RESUMO

Adipocytes are important but underappreciated components of bone marrow microenvironment, and their numbers greatly increase with age, obesity, and associated metabolic pathologies. Age and obesity are also significant risk factors for development of metastatic prostate cancer. Adipocytes are metabolically active cells that secrete adipokines, growth factors, and inflammatory mediators; influence behavior and function of neighboring cells; and have a potential to disturb local milleu and dysregulate normal bone homeostasis. Increased marrow adiposity has been linked to bone marrow inflammation and osteoporosis of the bone, but its effects on growth and progression of prostate tumors that have metastasized to the skeleton are currently not known. This review focuses on fat-bone relationship in a context of normal bone homeostasis and metastatic tumor growth in bone. We discuss effects of marrow fat cells on bone metabolism, hematopoiesis, and inflammation. Special attention is given to CCL2- and COX-2-driven pathways and their potential as therapeutic targets for bone metastatic disease.


Assuntos
Adipócitos/metabolismo , Medula Óssea/metabolismo , Medula Óssea/patologia , Adiposidade , Animais , Antineoplásicos/farmacologia , Antineoplásicos/uso terapêutico , Medula Óssea/efeitos dos fármacos , Neoplasias Ósseas/tratamento farmacológico , Neoplasias Ósseas/metabolismo , Neoplasias Ósseas/secundário , Osso e Ossos/metabolismo , Osso e Ossos/patologia , Homeostase , Humanos , Inflamação/metabolismo , Masculino , Neoplasias da Próstata/tratamento farmacológico , Neoplasias da Próstata/metabolismo , Neoplasias da Próstata/patologia , Transdução de Sinais/efeitos dos fármacos , Microambiente Tumoral
10.
Oncotarget ; 4(11): 2108-23, 2013 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-24240026

RESUMO

Incidence of skeletal metastases and death from prostate cancer greatly increases with age and obesity, conditions which increase marrow adiposity. Bone marrow adipocytes are metabolically active components of bone metastatic niche that modulate the function of neighboring cells; yet the mechanisms of their involvement in tumor behavior in bone have not been explored. In this study, using experimental models of intraosseous tumor growth and diet-induced obesity, we demonstrate the promoting effects of marrow fat on growth and progression of skeletal prostate tumors. We reveal that exposure to lipids supplied by marrow adipocytes induces expression of lipid chaperone FABP4, pro-inflammatory interleukin IL-1ß, and oxidative stress protein HMOX-1 in metastatic tumor cells and stimulates their growth and invasiveness. We show that FABP4 is highly overexpressed in prostate skeletal tumors from obese mice and in bone metastasis samples from prostate cancer patients. In addition, we provide results suggestive of bi-directional interaction between FABP4 and PPARγ pathways that may be driving aggressive tumor cell behavior in bone. Together, our data provide evidence for functional relationship between bone marrow adiposity and metastatic prostate cancers and unravel the FABP4/IL-1ß axis as a potential therapeutic target for this presently incurable disease.


Assuntos
Adipócitos/metabolismo , Medula Óssea/metabolismo , Neoplasias Ósseas/metabolismo , Neoplasias Ósseas/secundário , Proteínas de Ligação a Ácido Graxo/metabolismo , Neoplasias da Próstata/metabolismo , Neoplasias da Próstata/patologia , Adipócitos/patologia , Animais , Medula Óssea/patologia , Neoplasias Ósseas/genética , Processos de Crescimento Celular/fisiologia , Linhagem Celular Tumoral , Proteínas de Ligação a Ácido Graxo/genética , Humanos , Interleucina-1beta/metabolismo , Masculino , Camundongos , PPAR gama/metabolismo , Neoplasias da Próstata/genética , Transfecção , Células Tumorais Cultivadas , Regulação para Cima
11.
DNA Repair (Amst) ; 9(5): 579-87, 2010 May 04.
Artigo em Inglês | MEDLINE | ID: mdl-20338830

RESUMO

Human APOBEC3G (A3G) and activation-induced deaminase (AID) belong to a family of DNA-cytosine deaminases. While A3G targets the last C in a run of C's, AID targets C in the consensus sequence WRC (W is A or T and R is a purine). Guided by the structures of the A3G carboxyl-terminal catalytic domain (A3G-CTD), we identified two potential regions (region 1 and region 2) that may interact with DNA and swapped the corresponding regions between a variant of A3G-CTD and AID. The resulting hybrids were expressed in Escherichia coli and two different genetic assays and a biochemical assay were used to determine the sequence selectivity of the hybrids in promoting C to T mutations. The results show that while the 10 amino acid region 2 of A3G was its principal sequence-specificity determinant, region 1 of A3G enhanced the target cytosine preference conferred by region 2. In contrast, neither of the two regions in AID individually or in combination were sufficient to confer the DNA sequence preference of this protein upon A3G. Instead, introduction of AID sequences in A3G relaxed the sequence-specificity of the latter protein. Our results show that the sequence selectivity of APOBEC family of enzymes is determined by at least two separate sequence segments and there may be additional regions of the protein involved in DNA sequence recognition.


Assuntos
Citidina Desaminase/metabolismo , Desaminase APOBEC-3G , Sequência de Aminoácidos , Sequência de Bases , Citidina Desaminase/química , Citidina Desaminase/genética , DNA/genética , DNA/metabolismo , Humanos , Modelos Moleculares , Dados de Sequência Molecular , Mutação , Conformação Proteica , Proteínas Recombinantes de Fusão/química , Proteínas Recombinantes de Fusão/genética , Proteínas Recombinantes de Fusão/metabolismo , Especificidade por Substrato
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA