Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 30
Filtrar
1.
Case Rep Oncol Med ; 2024: 4756335, 2024.
Artigo em Inglês | MEDLINE | ID: mdl-38239272

RESUMO

Metastatic disease to the breast is a rare event, accounting for 0.5-2% of all breast cancers. Outside of metastases from the contralateral breast, malignant ovarian epithelial tumors are the most common origin of these metastases. Here, we present a very rare case of a high-grade ovarian serous adenocarcinoma presenting clinically as inflammatory breast cancer in a 70-year-old woman.

2.
J Clin Invest ; 133(6)2023 03 15.
Artigo em Inglês | MEDLINE | ID: mdl-36919699

RESUMO

High mobility group A1 (HMGA1) chromatin regulators are upregulated in diverse tumors where they portend adverse outcomes, although how they function in cancer remains unclear. Pancreatic ductal adenocarcinomas (PDACs) are highly lethal tumors characterized by dense desmoplastic stroma composed predominantly of cancer-associated fibroblasts and fibrotic tissue. Here, we uncover an epigenetic program whereby HMGA1 upregulates FGF19 during tumor progression and stroma formation. HMGA1 deficiency disrupts oncogenic properties in vitro while impairing tumor inception and progression in KPC mice and subcutaneous or orthotopic models of PDAC. RNA sequencing revealed HMGA1 transcriptional networks governing proliferation and tumor-stroma interactions, including the FGF19 gene. HMGA1 directly induces FGF19 expression and increases its protein secretion by recruiting active histone marks (H3K4me3, H3K27Ac). Surprisingly, disrupting FGF19 via gene silencing or the FGFR4 inhibitor BLU9931 recapitulates most phenotypes observed with HMGA1 deficiency, decreasing tumor growth and formation of a desmoplastic stroma in mouse models of PDAC. In human PDAC, overexpression of HMGA1 and FGF19 defines a subset of tumors with extremely poor outcomes. Our results reveal what we believe is a new paradigm whereby HMGA1 and FGF19 drive tumor progression and stroma formation, thus illuminating FGF19 as a rational therapeutic target for a molecularly defined PDAC subtype.


Assuntos
Carcinoma Ductal Pancreático , Neoplasias Pancreáticas , Animais , Humanos , Camundongos , Carcinogênese/genética , Carcinoma Ductal Pancreático/genética , Carcinoma Ductal Pancreático/patologia , Linhagem Celular Tumoral , Proliferação de Células , Fatores de Crescimento de Fibroblastos/genética , Fatores de Crescimento de Fibroblastos/metabolismo , Inativação Gênica , Proteína HMGA1a/genética , Proteína HMGA1a/metabolismo , Neoplasias Pancreáticas/patologia
3.
Cell Mol Gastroenterol Hepatol ; 15(4): 1000-1015, 2023.
Artigo em Inglês | MEDLINE | ID: mdl-36592863

RESUMO

BACKGROUND & AIMS: The adipokine CTRP3 has anti-inflammatory effects in several nonintestinal disorders. Although serum CTRP3 is reduced in patients with inflammatory bowel disease (IBD), its function in IBD has not been established. Here, we elucidate the function of CTRP3 in intestinal inflammation. METHODS: CTRP3 knockout (KO) and overexpressing transgenic (Tg) mice, along with their corresponding wild-type littermates, were treated with dextran sulfate sodium for 6-10 days. Colitis phenotypes and histologic data were analyzed. CTRP3-mediated signaling was examined in murine and human intestinal mucosa and mouse intestinal organoids derived from CTRP3 KO and Tg mice. RESULTS: CTRP3 KO mice developed more severe colitis, whereas CTRP3 Tg mice developed less severe colitis than wild-type littermates. The deletion of CTRP3 correlated with decreased levels of Sirtuin-1 (SIRT1), a histone deacetylase, and increased levels of phosphorylated/acetylated NF-κB subunit p65 and proinflammatory cytokines tumor necrosis factor-α and interleukin-6. Results from CTRP3 Tg mice were inverse to those from CTRP3 KO mice. The addition of SIRT1 activator resveratrol to KO intestinal organoids and SIRT1 inhibitor Ex-527 to Tg intestinal organoids suggest that SIRT1 is a downstream effector of CTRP3-related inflammatory changes. In patients with IBD, a similar CTRP3/SIRT1/NF-κB relationship was observed. CONCLUSIONS: CTRP3 expression levels correlate negatively with intestinal inflammation in acute mouse colitis models and patients with IBD. CTRP3 may attenuate intestinal inflammation via SIRT1/NF-κB signaling. The manipulation of CTRP3 signaling, including through the use of SIRT1 activators, may offer translational potential in the treatment of IBD.


Assuntos
Colite , Doenças Inflamatórias Intestinais , Animais , Humanos , Camundongos , Adipocinas/metabolismo , Colite/induzido quimicamente , Colite/metabolismo , Complemento C1q , Inflamação/metabolismo , Camundongos Knockout , Camundongos Transgênicos , NF-kappa B/metabolismo , Sirtuína 1/genética , Sirtuína 1/metabolismo , Fator de Necrose Tumoral alfa
4.
Blood ; 139(18): 2797-2815, 2022 05 05.
Artigo em Inglês | MEDLINE | ID: mdl-35286385

RESUMO

Myeloproliferative neoplasms (MPNs) transform to myelofibrosis (MF) and highly lethal acute myeloid leukemia (AML), although the actionable mechanisms driving progression remain elusive. Here, we elucidate the role of the high mobility group A1 (HMGA1) chromatin regulator as a novel driver of MPN progression. HMGA1 is upregulated in MPN, with highest levels after transformation to MF or AML. To define HMGA1 function, we disrupted gene expression via CRISPR/Cas9, short hairpin RNA, or genetic deletion in MPN models. HMGA1 depletion in JAK2V617F AML cell lines disrupts proliferation, clonogenicity, and leukemic engraftment. Surprisingly, loss of just a single Hmga1 allele prevents progression to MF in JAK2V617F mice, decreasing erythrocytosis, thrombocytosis, megakaryocyte hyperplasia, and expansion of stem and progenitors, while preventing splenomegaly and fibrosis within the spleen and BM. RNA-sequencing and chromatin immunoprecipitation sequencing revealed HMGA1 transcriptional networks and chromatin occupancy at genes that govern proliferation (E2F, G2M, mitotic spindle) and cell fate, including the GATA2 master regulatory gene. Silencing GATA2 recapitulates most phenotypes observed with HMGA1 depletion, whereas GATA2 re-expression partially rescues leukemogenesis. HMGA1 transactivates GATA2 through sequences near the developmental enhancer (+9.5), increasing chromatin accessibility and recruiting active histone marks. Further, HMGA1 transcriptional networks, including proliferation pathways and GATA2, are activated in human MF and MPN leukemic transformation. Importantly, HMGA1 depletion enhances responses to the JAK2 inhibitor, ruxolitinib, preventing MF and prolonging survival in murine models of JAK2V617F AML. These findings illuminate HMGA1 as a key epigenetic switch involved in MPN transformation and a promising therapeutic target to treat or prevent disease progression.


Assuntos
Fator de Transcrição GATA2 , Proteína HMGA1a , Leucemia Mieloide Aguda , Transtornos Mieloproliferativos , Mielofibrose Primária , Animais , Proliferação de Células , Cromatina/genética , Fator de Transcrição GATA2/genética , Redes Reguladoras de Genes , Proteína HMGA1a/genética , Proteína HMGA1a/metabolismo , Janus Quinase 2/genética , Janus Quinase 2/metabolismo , Leucemia Mieloide Aguda/genética , Camundongos , Transtornos Mieloproliferativos/genética , Transtornos Mieloproliferativos/metabolismo , Mielofibrose Primária/genética
5.
Breast Cancer Res Treat ; 179(1): 25-35, 2020 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-31531802

RESUMO

PURPOSE: The high mobility group A1 (HMGA1) chromatin remodeling protein is required for metastatic progression and cancer stem cell properties in preclinical breast cancer models, although its role in breast carcinogenesis has remained unclear. To investigate HMGA1 in primary breast cancer, we evaluated immunoreactivity score (IRS) in tumors from a large cohort of Asian women; HMGA1 gene expression was queried from two independent Western cohorts. METHODS: HMGA1 IRS was generated from breast tumors in Korean women as the product of staining intensity (weak = 1, moderate = 2, strong = 3) and percent positive cells (< 5% = 0, 5-30% = 1, 30-60% = 2, > 60% = 3), and stratified into three groups: low (< 3), intermediate (3-6), high (> 6). We assessed HMGA1 and estrogen receptor (ESR1) gene expression from two large databases (TCGA, METABRIC). Overall survival was ascertained from the METABRIC cohort. RESULTS: Among 540 primary tumors from Korean women (181 ER-negative, 359 ER-positive), HMGA1 IRS was < 3 in 89 (16.5%), 3-6 in 215 (39.8%), and > 6 in 236 (43.7%). High HMGA1 IRS was associated with estrogen receptor (ER)-negativity (χ2 = 12.07; P = 0.002) and advanced nuclear grade (χ2 = 12.83; P = 0.012). In two large Western cohorts, the HMGA1 gene was overexpressed in breast cancers compared to non-malignant breast tissue (P < 0.0001), including Asian, African American, and Caucasian subgroups. HMGA1 was highest in ER-negative tumors and there was a strong inverse correlation between HMGA1 and ESR1 gene expression (Pearson r = - 0.60, P < 0.0001). Most importantly, high HMGA1 predicted decreased overall survival (P < 0.0001) for all women with breast cancer and further stratified ER-positive tumors into those with inferior outcomes. CONCLUSIONS: Together, our results suggest that HMGA1 contributes to estrogen-independence, tumor progression, and poor outcomes. Moreover, further studies are warranted to determine whether HMGA1 could serve as a prognostic marker and therapeutic target for women with breast cancer.


Assuntos
Neoplasias da Mama/metabolismo , Proteína HMGA1a/genética , Proteína HMGA1a/metabolismo , Receptores de Estrogênio/metabolismo , Adulto , Idoso , Idoso de 80 Anos ou mais , Neoplasias da Mama/genética , Progressão da Doença , Feminino , Humanos , Pessoa de Meia-Idade , Prognóstico , República da Coreia , Análise de Sobrevida , Regulação para Cima , Adulto Jovem
6.
J Vis Exp ; (147)2019 05 10.
Artigo em Inglês | MEDLINE | ID: mdl-31132065

RESUMO

Intestinal organoid cultures provide a unique opportunity to investigate intestinal stem cell and crypt biology in vitro, although efficient approaches to manipulate gene expression in organoids have made limited progress in this arena. While CRISPR/Cas9 technology allows for precise genome editing of cells for organoid generation, this strategy requires extensive selection and screening by sequence analysis, which is both time-consuming and costly. Here, we provide a detailed protocol for efficient viral transduction of intestinal organoids. This approach is rapid and highly efficient, thus decreasing the time and expense inherent in CRISPR/Cas9 technology. We also present a protocol to generate frozen sections from intact organoid cultures for further analysis with immunohistochemical or immunofluorescent staining, which can be used to confirm gene expression or silencing. After successful transduction of viral vectors for gene expression or silencing is achieved, intestinal stem cell and crypt function can be rapidly assessed. Although most organoid studies employ in vitro assays, organoids can also be delivered to mice for in vivo functional analyses. Moreover, our approaches are advantageous for predicting therapeutic responses to drugs because currently available therapies generally function by modulating gene expression or protein function rather than altering the genome.


Assuntos
Secções Congeladas , Engenharia Genética/métodos , Vetores Genéticos/metabolismo , Intestinos/fisiologia , Nanopartículas de Magnetita/química , Organoides/metabolismo , Transdução Genética , Animais , DNA/genética , Edição de Genes/métodos , Células HEK293 , Humanos , Campos Magnéticos , Camundongos
7.
Cancer Res ; 78(8): 1890-1897, 2018 04 15.
Artigo em Inglês | MEDLINE | ID: mdl-29618461

RESUMO

High mobility group A1 (HMGA1) chromatin remodeling proteins are enriched in aggressive cancers and stem cells, although their common function in these settings has remained elusive until now. Recent work in murine intestinal stem cells (ISC) revealed a novel role for Hmga1 in enhancing self-renewal by amplifying Wnt signaling, both by inducing genes expressing Wnt agonist receptors and Wnt effectors. Surprisingly, Hmga1 also "builds" a stem cell niche by upregulating Sox9, a factor required for differentiation to Paneth cells; these cells constitute an epithelial niche by secreting Wnt and other factors to support ISCs. HMGA1 is also highly upregulated in colon cancer compared with nonmalignant epithelium and SOX9 becomes overexpressed during colon carcinogenesis. Intriguingly, HMGA1 is overexpressed in diverse cancers with poor outcomes, where it regulates developmental genes. Similarly, HMGA1 induces genes responsible for pluripotency and self-renewal in embryonic stem cells. These findings demonstrate that HMGA1 maintains Wnt and other developmental transcriptional networks and suggest that HMGA1 overexpression fosters carcinogenesis and tumor progression through dysregulation of these pathways. Studies are now needed to determine more precisely how HMGA1 modulates chromatin structure to amplify developmental genes and how to disrupt this process in cancer therapy. Cancer Res; 78(8); 1890-7. ©2018 AACR.


Assuntos
Neoplasias Colorretais/patologia , Proteína HMGA1a/metabolismo , Células-Tronco/metabolismo , Proteínas Wnt/metabolismo , Animais , Carcinogênese , Diferenciação Celular , Linhagem da Célula , Neoplasias Colorretais/metabolismo , Progressão da Doença , Desenvolvimento Embrionário , Células-Tronco Embrionárias/metabolismo , Proteína HMGA1a/genética , Humanos , Nicho de Células-Tronco
8.
Bone Res ; 6: 5, 2018.
Artigo em Inglês | MEDLINE | ID: mdl-29507819

RESUMO

Parathyroid hormone (PTH) regulates bone remodeling by activating PTH type 1 receptor (PTH1R) in osteoblasts/osteocytes. Insulin-like growth factor type 1 (IGF-1) stimulates mesenchymal stem cell differentiation to osteoblasts. However, little is known about the signaling mechanisms that regulates the osteoblast-to-osteocyte transition. Here we report that PTH and IGF-I synergistically enhance osteoblast-to-osteocyte differentiation. We identified that a specific tyrosine residue, Y494, on the cytoplasmic domain of PTH1R can be phosphorylated by insulin-like growth factor type I receptor (IGF1R) in vitro. Phosphorylated PTH1R localized to the barbed ends of actin filaments and increased actin polymerization during morphological change of osteoblasts into osteocytes. Disruption of the phosphorylation site reduced actin polymerization and dendrite length. Mouse models with conditional ablation of PTH1R in osteoblasts demonstrated a reduction in the number of osteoctyes and dendrites per osteocyte, with complete overlap of PTH1R with phosphorylated-PTH1R positioning in osteocyte dendrites in wild-type mice. Thus, our findings reveal a novel signaling mechanism that enhances osteoblast-to-osteocyte transition by direct phosphorylation of PTH1R by IGF1R.

9.
Nat Commun ; 8: 15008, 2017 04 28.
Artigo em Inglês | MEDLINE | ID: mdl-28452345

RESUMO

High-mobility group A1 (Hmga1) chromatin remodelling proteins are enriched in intestinal stem cells (ISCs), although their function in this setting was unknown. Prior studies showed that Hmga1 drives hyperproliferation, aberrant crypt formation and polyposis in transgenic mice. Here we demonstrate that Hmga1 amplifies Wnt/ß-catenin signalling to enhance self-renewal and expand the ISC compartment. Hmga1 upregulates genes encoding both Wnt agonist receptors and downstream Wnt effectors. Hmga1 also helps to 'build' an ISC niche by expanding the Paneth cell compartment and directly inducing Sox9, which is required for Paneth cell differentiation. In human intestine, HMGA1 and SOX9 are positively correlated, and both become upregulated in colorectal cancer. Our results define a unique role for Hmga1 in intestinal homeostasis by maintaining the stem cell pool and fostering terminal differentiation to establish an epithelial stem cell niche. This work also suggests that deregulated Hmga1 perturbs this equilibrium during intestinal carcinogenesis.


Assuntos
Proteína HMGA1a/metabolismo , Mucosa Intestinal/metabolismo , Celulas de Paneth/metabolismo , Células-Tronco/metabolismo , Via de Sinalização Wnt , Animais , Diferenciação Celular/genética , Proliferação de Células/genética , Células Cultivadas , Proteína HMGA1a/genética , Humanos , Mucosa Intestinal/citologia , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Microscopia Confocal , Celulas de Paneth/citologia , Fatores de Transcrição SOX9/genética , Fatores de Transcrição SOX9/metabolismo , Nicho de Células-Tronco , Células-Tronco/citologia , Imagem com Lapso de Tempo
11.
J Proteome Res ; 15(12): 4176-4187, 2016 12 02.
Artigo em Inglês | MEDLINE | ID: mdl-27696867

RESUMO

Because colorectal cancer (CRC) remains a leading cause of cancer mortality worldwide, more accessible screening tests are urgently needed to identify early stage lesions. We hypothesized that highly sensitive, metabolic profile analysis of stool samples will identify metabolites associated with early stage lesions and could serve as a noninvasive screening test. We therefore applied traveling wave ion mobility mass spectrometry (TWIMMS) coupled with ultraperformance liquid chromatography (UPLC) to investigate metabolic aberrations in stool samples in a transgenic model of premalignant polyposis aberrantly expressing the gene encoding the high mobility group A (Hmga1) chromatin remodeling protein. Here, we report for the first time that the fecal metabolome of Hmga1 mice is distinct from that of control mice and includes metabolites previously identified in human CRC. Significant alterations were observed in fatty acid metabolites and metabolites associated with bile acids (hypoxanthine xanthine, taurine) in Hmga1 mice compared to controls. Surprisingly, a marked increase in the levels of distinctive short, arginine-enriched, tetra-peptide fragments was observed in the transgenic mice. Together these findings suggest that specific metabolites are associated with Hmga1-induced polyposis and abnormal proliferation in intestinal epithelium. Although further studies are needed, these data provide a compelling rationale to develop fecal metabolomic analysis as a noninvasive screening tool to detect early precursor lesions to CRC in humans.


Assuntos
Neoplasias Colorretais/diagnóstico , Neoplasias Colorretais/metabolismo , Detecção Precoce de Câncer/métodos , Fezes/química , Proteínas HMGA/genética , Metaboloma , Polipose Adenomatosa do Colo/genética , Animais , Ácidos e Sais Biliares/metabolismo , Cromatografia Líquida de Alta Pressão , Neoplasias Colorretais/patologia , Modelos Animais de Doenças , Ácidos Graxos/metabolismo , Espectrometria de Massas , Camundongos , Camundongos Transgênicos , Fragmentos de Peptídeos/metabolismo
12.
Leuk Lymphoma ; 57(11): 2681-4, 2016 11.
Artigo em Inglês | MEDLINE | ID: mdl-26952843
13.
Gynecol Oncol ; 141(3): 580-587, 2016 06.
Artigo em Inglês | MEDLINE | ID: mdl-27001612

RESUMO

OBJECTIVES: Although uterine cancer is the fourth most common cause for cancer death in women worldwide, the molecular underpinnings of tumor progression remain poorly understood. The High Mobility Group A1 (HMGA1) gene is overexpressed in aggressive cancers and high levels portend adverse outcomes in diverse tumors. We previously reported that Hmga1a transgenic mice develop uterine tumors with complete penetrance. Because HMGA1 drives tumor progression by inducing MatrixMetalloproteinase (MMP) and other genes involved in invasion, we explored the HMGA1-MMP-2 pathway in uterine cancer. METHODS: To investigate MMP-2 in uterine tumors driven by HMGA1, we used a genetic approach with mouse models. Next, we assessed HMGA1 and MMP-2 expression in primary human uterine tumors, including low-grade carcinomas (endometrial endometrioid) and more aggressive tumors (endometrial serous carcinomas, uterine carcinosarcomas/malignant mesodermal mixed tumors). RESULTS: Here, we report for the first time that uterine tumor growth is impaired in Hmga1a transgenic mice crossed on to an Mmp-2 deficient background. In human tumors, we discovered that HMGA1 is highest in aggressive carcinosarcomas and serous carcinomas, with lower levels in the more indolent endometrioid carcinomas. Moreover, HMGA1 and MMP-2 were positively correlated, but only in a subset of carcinosarcomas. HMGA1 also occupies the MMP-2 promoter in human carcinosarcoma cells. CONCLUSIONS: Together, our studies define a novel HMGA1-MMP-2 pathway involved in a subset of human carcinosarcomas and tumor progression in murine models. Our work also suggests that targeting HMGA1 could be effective adjuvant therapy for more aggressive uterine cancers and provides compelling data for further preclinical studies.


Assuntos
Carcinossarcoma/genética , Cistadenocarcinoma Seroso/genética , Proteína HMGA1a/genética , Metaloproteinase 2 da Matriz/genética , Neoplasias Uterinas/genética , Animais , Carcinossarcoma/metabolismo , Imunoprecipitação da Cromatina , Cistadenocarcinoma Seroso/metabolismo , Feminino , Inativação Gênica , Proteína HMGA1a/biossíntese , Humanos , Masculino , Metaloproteinase 2 da Matriz/biossíntese , Camundongos Transgênicos , Regiões Promotoras Genéticas , Regulação para Cima , Neoplasias Uterinas/metabolismo
14.
Methods Mol Biol ; 1344: 287-300, 2016.
Artigo em Inglês | MEDLINE | ID: mdl-26520132

RESUMO

TGF-ß signaling plays a key role in the temporal and spatial regulation of bone remodeling. During osteoclast bone resorption, TGF-ß is released from the bone matrix and activated. Active TGF-ß recruits mesenchymal stem cells to the bone resorption pit through the SMAD signaling pathway. Mesenchymal stem cells undergo osteoblast differentiation and deposit new bone filling in the resorption pit and maintaining the structural integrity of the skeleton. Thus, TGF-ß signaling plays a key role in the coupling process and disruptions to the TGF-ß signaling pathway lead to loss of skeletal integrity. This chapter describes methods on how to quantitate bone matrix TGF-ß and assess its role in mesenchymal stem cell migration both in vitro and in vivo.


Assuntos
Remodelação Óssea , Transdução de Sinais , Fator de Crescimento Transformador beta/metabolismo , Animais , Medula Óssea/metabolismo , Remodelação Óssea/genética , Movimento Celular/genética , Matriz Extracelular/metabolismo , Imuno-Histoquímica , Técnicas In Vitro , Camundongos , Ratos , Fator de Crescimento Transformador beta/genética
15.
Appl Microbiol Biotechnol ; 99(19): 8035-43, 2015 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-26048470

RESUMO

Neuritin (also known as candidate plasticity gene 15 (cpg15)) is a neurotrophic factor that was recently discovered in a screen aimed at identifying genes involved in activity-dependent synaptic plasticity. Neuritin plays multiple roles in both neural development (Chen et al. Zhonghua Yan Ke Za Zhi 46:978-983 2010; Corriveau et al. J Neurosci 19:7999-8008 1999; Lee and Nedivi J Neurosci 22:1807-1815 2002) and synaptic plasticity (Fujino et al. Gene Dev 25:2674-2685 2011; Leslie and Nedivi Prog 14 Neurobiol 94:223-237 2011; Loebrich and Nedivi Physiol Rev 89:1079 2009). In this study, to produce bioactive, soluble recombinant human neuritin protein, a portion of NRN1 was cloned into the Pichia pastoris expression vector pPIC9K. The recombinant vector was then transformed into the methylotrophic yeast strain P. pastoris GS115, and a shaking flask method and His-tag purification strategy were utilized to express and purify neuritin protein. The resulting protein had a molecular mass of approximately 11 kDa, and subsequent functional analyses indicated that the purified neuritin promoted neurite outgrowth from embryonic chicken dorsal root ganglions, while also prolonging the survival of these ganglions, and from PC12 cells. These findings suggest that neuritin was expressed effectively in vitro and that this protein may play a role in stimulating neurite outgrowth of both dorsal root ganglions and PC12 cells. This study provides a novel strategy for the large-scale production of bioactive neuritin, which will enable further study of the biological function of this protein.


Assuntos
Fatores de Crescimento Neural/genética , Fatores de Crescimento Neural/isolamento & purificação , Neuropeptídeos/genética , Neuropeptídeos/isolamento & purificação , Pichia/genética , Animais , Células Cultivadas , Proteínas Ligadas por GPI/genética , Proteínas Ligadas por GPI/isolamento & purificação , Proteínas Ligadas por GPI/metabolismo , Proteínas Ligadas por GPI/farmacologia , Expressão Gênica , Humanos , Fatores de Crescimento Neural/metabolismo , Fatores de Crescimento Neural/farmacologia , Neuritos/efeitos dos fármacos , Neuropeptídeos/metabolismo , Neuropeptídeos/farmacologia , Pichia/metabolismo , Ratos , Proteínas Recombinantes/genética , Proteínas Recombinantes/isolamento & purificação , Proteínas Recombinantes/metabolismo , Proteínas Recombinantes/farmacologia
17.
J Proteome Res ; 14(3): 1420-31, 2015 Mar 06.
Artigo em Inglês | MEDLINE | ID: mdl-25643065

RESUMO

Although significant progress has been made in the diagnosis and treatment of colorectal cancer (CRC), it remains a leading cause of cancer death worldwide. Early identification and removal of polyps that may progress to overt CRC is the cornerstone of CRC prevention. Expression of the High Mobility Group A1 (HMGA1) gene is significantly elevated in CRCs as compared with adjacent, nonmalignant tissues. We investigated metabolic aberrations induced by HMGA1 overexpression in small intestinal and colonic epithelium using traveling wave ion mobility mass spectrometry (TWIMMS) in a transgenic model in which murine Hmga1 was misexpressed in colonic epithelium. To determine if these Hmga1-induced metabolic alterations in mice were relevant to human colorectal carcinogenesis, we also investigated tumors from patients with CRC and matched, adjacent, nonmalignant tissues. Multivariate statistical methods and manual comparisons were used to identify metabolites specific to Hmga1 and CRC. Statistical modeling of data revealed distinct metabolic patterns in Hmga1 transgenics and human CRC samples as compared with the control tissues. We discovered that 13 metabolites were specific for Hmga1 in murine intestinal epithelium and also found in human CRC. Several of these metabolites function in fatty acid metabolism and membrane composition. Although further validation is needed, our results suggest that high levels of HMGA1 protein drive metabolic alterations that contribute to CRC pathogenesis through fatty acid synthesis. These metabolites could serve as potential biomarkers or therapeutic targets.


Assuntos
Polipose Adenomatosa do Colo/fisiopatologia , Proliferação de Células/fisiologia , Neoplasias Colorretais/patologia , Proteína HMGA1a/fisiologia , Mucosa Intestinal/patologia , Neoplasias Colorretais/metabolismo , Proteína HMGA1a/metabolismo , Humanos , Mucosa Intestinal/metabolismo , Espectrometria de Massas em Tandem
18.
J Bone Miner Res ; 30(2): 309-17, 2015 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-25196529

RESUMO

Longitudinal growth of postnatal bone requires precise control of growth plate cartilage chondrocytes and subsequent osteogenesis and bone formation. Little is known about the role of angiogenesis and bone remodeling in maintenance of cartilaginous growth plate. Parathyroid hormone (PTH) stimulates bone remodeling by activating PTH receptor (PTH1R). Mice with conditional deletion of PTH1R in osteoblasts showed disrupted trabecular bone formation. The mice also exhibited postnatal growth retardation with profound defects in growth plate cartilage, ascribable predominantly to a decrease in number of hypertrophic chondrocytes, resulting in premature fusion of the growth plate and shortened long bones. Further characterization of hypertrophic zone and primary spongiosa revealed that endochondral angiogenesis and vascular invasion of the cartilage were impaired, which was associated with aberrant chondrocyte maturation and cartilage development. These studies reveal that PTH1R signaling in osteoblasts regulates cartilaginous growth plate for postnatal growth of bone.


Assuntos
Lâmina de Crescimento/irrigação sanguínea , Lâmina de Crescimento/crescimento & desenvolvimento , Lâmina de Crescimento/metabolismo , Neovascularização Fisiológica , Osteoblastos/metabolismo , Receptor Tipo 1 de Hormônio Paratireóideo/metabolismo , Transdução de Sinais , Animais , Animais Recém-Nascidos , Contagem de Células , Condrócitos/metabolismo , Condrócitos/patologia , Regulação da Expressão Gênica no Desenvolvimento , Hipertrofia , Camundongos Knockout , Osteogênese , Receptor Tipo 1 de Hormônio Paratireóideo/deficiência
19.
Nat Med ; 20(11): 1270-8, 2014 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-25282358

RESUMO

Osteogenesis during bone modeling and remodeling is coupled with angiogenesis. A recent study showed that a specific vessel subtype, strongly positive for CD31 and endomucin (CD31(hi)Emcn(hi)), couples angiogenesis and osteogenesis. Here, we found that platelet-derived growth factor-BB (PDGF-BB) secreted by preosteoclasts induces CD31(hi)Emcn(hi) vessel formation during bone modeling and remodeling. Mice with depletion of PDGF-BB in the tartrate-resistant acid phosphatase-positive cell lineage show significantly lower trabecular and cortical bone mass, serum and bone marrow PDGF-BB concentrations, and fewer CD31(hi)Emcn(hi) vessels compared to wild-type mice. In the ovariectomy (OVX)-induced osteoporotic mouse model, serum and bone marrow levels of PDGF-BB and numbers of CD31(hi)Emcn(hi) vessels are significantly lower compared to sham-operated controls. Treatment with exogenous PDGF-BB or inhibition of cathepsin K to increase the number of preosteoclasts, and thus the endogenous levels of PDGF-BB, increases CD31(hi)Emcn(hi) vessel number and stimulates bone formation in OVX mice. Thus, pharmacotherapies that increase PDGF-BB secretion from preosteoclasts offer a new therapeutic target for treating osteoporosis by promoting angiogenesis and thus bone formation.


Assuntos
Neovascularização Fisiológica , Osteoclastos/metabolismo , Osteogênese , Proteínas Proto-Oncogênicas c-sis/metabolismo , Fosfatase Ácida/metabolismo , Animais , Becaplermina , Catepsina K/antagonistas & inibidores , Catepsina K/metabolismo , Contagem de Células , Movimento Celular/efeitos dos fármacos , Meios de Cultivo Condicionados/farmacologia , Células Endoteliais/citologia , Células Endoteliais/efeitos dos fármacos , Células Endoteliais/metabolismo , Feminino , Fêmur/diagnóstico por imagem , Fêmur/efeitos dos fármacos , Fêmur/metabolismo , Proteína-Tirosina Quinases de Adesão Focal/metabolismo , Isoenzimas/metabolismo , Células-Tronco Mesenquimais/citologia , Células-Tronco Mesenquimais/efeitos dos fármacos , Células-Tronco Mesenquimais/metabolismo , Camundongos Endogâmicos C57BL , Neovascularização Fisiológica/efeitos dos fármacos , Osteoclastos/efeitos dos fármacos , Osteoclastos/enzimologia , Osteogênese/efeitos dos fármacos , Ovariectomia , Fosforilação/efeitos dos fármacos , Molécula-1 de Adesão Celular Endotelial a Plaquetas/metabolismo , Inibidores de Proteases/farmacologia , Proteínas Proto-Oncogênicas c-akt/metabolismo , Fosfatase Ácida Resistente a Tartarato , Microtomografia por Raio-X
20.
Mol Cell Biochem ; 397(1-2): 235-43, 2014 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-25239147

RESUMO

Stem cells dwell at the "stem cell niche" to accomplish a series of biological processes. The composition of the niche should be determined because the insufficient understanding of this feature limits the development in the study of stem cells. We showed in our study on mesenchymal stem cells (MSCs) that the MSCs first neighbored to CD31(+) cells, which proved to be endothelial progenitor cells (EPCs), and formed a group of cell colony before they exerted their biological functions. It was further proved that EPCs have close interactions with MSCs and promoted the self-renewal of the MSCs in vitro and in vivo. Together with these achievements, we hypothesized that EPCs may be a possible biological component of the MSC stem cell niche and affect the biological processes of MSCs.


Assuntos
Células Endoteliais/metabolismo , Células-Tronco Mesenquimais/metabolismo , Nicho de Células-Tronco/fisiologia , Animais , Células Endoteliais/citologia , Células-Tronco Mesenquimais/citologia , Camundongos , Camundongos Knockout
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...