Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 40
Filtrar
1.
Biochem Biophys Res Commun ; 710: 149881, 2024 May 28.
Artículo en Inglés | MEDLINE | ID: mdl-38583233

RESUMEN

Maackia amurensis lectins serve as research and botanical agents that bind to sialic residues on proteins. For example, M. amurensis seed lectin (MASL) targets the sialic acid modified podoplanin (PDPN) receptor to suppress arthritic chondrocyte inflammation, and inhibit tumor cell growth and motility. However, M. amurensis lectin nomenclature and composition are not clearly defined. Here, we sought to definitively characterize MASL and its effects on tumor cell behavior. We utilized SDS-PAGE and LC-MS/MS to find that M. amurensis lectins can be divided into two groups. MASL is a member of one group which is composed of subunits that form dimers, evidently mediated by a cysteine residue in the carboxy region of the protein. In contrast to MASL, members of the other group do not dimerize under nonreducing conditions. These data also indicate that MASL is composed of 4 isoforms with an identical amino acid sequence, but unique glycosylation sites. We also produced a novel recombinant soluble human PDPN receptor (shPDPN) with 17 threonine residues glycosylated with sialic acid moieties with potential to act as a ligand trap that inhibits OSCC cell growth and motility. In addition, we report here that MASL targets PDPN with very strong binding kinetics in the nanomolar range. Moreover, we confirm that MASL can inhibit the growth and motility of human oral squamous cell carcinoma (OSCC) cells that express the PDPN receptor. Taken together, these data characterize M. amurensis lectins into two major groups based on their intrinsic properties, clarify the composition of MASL and its subunit isoform sequence and glycosylation sites, define sialic acid modifications on the PDPN receptor and its ability to act as a ligand trap, quantitate MASL binding to PDPN with KD in the nanomolar range, and verify the ability of MASL to serve as a potential anticancer agent.


Asunto(s)
Antineoplásicos , Carcinoma de Células Escamosas , Neoplasias de Cabeza y Cuello , Neoplasias de la Boca , Humanos , Carcinoma de Células Escamosas/patología , Carcinoma de Células Escamosas de Cabeza y Cuello , Ácido N-Acetilneuramínico/metabolismo , Maackia/química , Maackia/metabolismo , Neoplasias de la Boca/patología , Cromatografía Liquida , Ligandos , Espectrometría de Masas en Tándem , Lectinas/farmacología , Antineoplásicos/farmacología , Análisis de Secuencia , Movimiento Celular
2.
Mol Carcinog ; 61(7): 677-689, 2022 07.
Artículo en Inglés | MEDLINE | ID: mdl-35472679

RESUMEN

The Src tyrosine kinase is a strong tumor promotor. Over a century of research has elucidated fundamental mechanisms that drive its oncogenic potential. Src phosphorylates effector proteins to promote hallmarks of tumor progression. For example, Src associates with the Cas focal adhesion adaptor protein to promote anchorage independent cell growth. In addition, Src phosphorylates Cas to induce Pdpn expression to promote cell migration. Pdpn is a transmembrane receptor that can independently increase cell migration in the absence of oncogenic Src kinase activity. However, to our knowledge, effects of Src kinase activity on anchorage independent cell growth and migration have not been examined in the absence of Pdpn expression. Here, we analyzed the effects of an inducible Src kinase construct in knockout cells with and without exogenous Pdpn expression on cell morphology migration and anchorage independent growth. We report that Src promoted anchorage independent cell growth in the absence of Pdpn expression. In contrast, Src was not able to promote cell migration in the absence of Pdpn expression. In addition, continued Src kinase activity was required for cells to assume a transformed morphology since cells reverted to a nontransformed morphology upon cessation of Src kinase activity. We also used phosphoproteomic analysis to identify 28 proteins that are phosphorylated in Src transformed cells in a Pdpn dependent manner. Taken together, these data indicate that Src utilizes Pdpn to promote transformed cell growth and motility in complementary, but parallel, as opposed to serial, pathways.


Asunto(s)
Neoplasias , Familia-src Quinasas , Adhesión Celular , Movimiento Celular , Humanos , Fosforilación , Familia-src Quinasas/genética , Familia-src Quinasas/metabolismo
3.
Cell Commun Signal ; 20(1): 19, 2022 02 17.
Artículo en Inglés | MEDLINE | ID: mdl-35177067

RESUMEN

BACKGROUND: The Src tyrosine kinase phosphorylates effector proteins to induce expression of the podoplanin (PDPN) receptor in order to promote tumor progression. However, nontransformed cells can normalize the growth and morphology of neighboring transformed cells. Transformed cells must escape this process, called "contact normalization", to become invasive and malignant. Contact normalization requires junctional communication between transformed and nontransformed cells. However, specific junctions that mediate this process have not been defined. This study aimed to identify junctional proteins required for contact normalization. METHODS: Src transformed cells and oral squamous cell carcinoma cells were cultured with nontransformed cells. Formation of heterocellular adherens junctions between transformed and nontransformed cells was visualized by fluorescent microscopy. CRISPR technology was used to produce cadherin deficient and cadherin competent nontransformed cells to determine the requirement for adherens junctions during contact normalization. Contact normalization of transformed cells cultured with cadherin deficient or cadherin competent nontransformed cells was analyzed by growth assays, immunofluorescence, western blotting, and RNA-seq. In addition, Src transformed cells expressing PDPN under a constitutively active exogenous promoter were used to examine the ability of PDPN to override contact normalization. RESULTS: We found that N-cadherin (N-Cdh) appeared to mediate contact normalization. Cadherin competent cells that expressed N-Cdh inhibited the growth of neighboring transformed cells in culture, while cadherin deficient cells failed to inhibit the growth of these cells. Results from RNA-seq analysis indicate that about 10% of the transcripts affected by contact normalization relied on cadherin mediated communication, and this set of genes includes PDPN. In contrast, cadherin deficient cells failed to inhibit PDPN expression or normalize the growth of adjacent transformed cells. These data indicate that nontransformed cells formed heterocellular cadherin junctions to inhibit PDPN expression in adjacent transformed cells. Moreover, we found that PDPN enabled transformed cells to override the effects of contact normalization in the face of continued N-Cdh expression. Cadherin competent cells failed to normalize the growth of transformed cells expressing PDPN under a constitutively active exogenous promoter. CONCLUSIONS: Nontransformed cells form cadherin junctions with adjacent transformed cells to decrease PDPN expression in order to inhibit tumor cell proliferation. Cancer begins when a single cell acquires changes that enables them to form tumors. During these beginning stages of cancer development, normal cells surround and directly contact the cancer cell to prevent tumor formation and inhibit cancer progression. This process is called contact normalization. Cancer cells must break free from contact normalization to progress into a malignant cancer. Contact normalization is a widespread and powerful process; however, not much is known about the mechanisms involved in this process. This work identifies proteins required to form contacts between normal cells and cancer cells, and explores pathways by which cancer cells override contact normalization to progress into malignant cancers. Video Abstract.


Asunto(s)
Antígenos CD , Cadherinas , Neoplasias de la Boca , Carcinoma de Células Escamosas de Cabeza y Cuello , Uniones Adherentes/metabolismo , Antígenos CD/metabolismo , Cadherinas/metabolismo , Transformación Celular Neoplásica , Humanos , Neoplasias de la Boca/metabolismo , Neoplasias de la Boca/patología , Carcinoma de Células Escamosas de Cabeza y Cuello/metabolismo , Carcinoma de Células Escamosas de Cabeza y Cuello/patología
4.
Exp Cell Res ; 403(1): 112594, 2021 06 01.
Artículo en Inglés | MEDLINE | ID: mdl-33823179

RESUMEN

COVID-19 was declared an international public health emergency in January, and a pandemic in March of 2020. There are over 125 million confirmed COVID-19 cases that have caused over 2.7 million deaths worldwide as of March 2021. COVID-19 is caused by the SARS-CoV-2 virus. SARS-CoV-2 presents a surface "spike" protein that binds to the ACE2 receptor to infect host cells. In addition to the respiratory tract, SARS-Cov-2 can also infect cells of the oral mucosa, which also express the ACE2 receptor. The spike and ACE2 proteins are highly glycosylated with sialic acid modifications that direct viral-host interactions and infection. Maackia amurensis seed lectin (MASL) has a strong affinity for sialic acid modified proteins and can be used as an antiviral agent. Here, we report that MASL targets the ACE2 receptor, decreases ACE2 expression and glycosylation, suppresses binding of the SARS-CoV-2 spike protein, and decreases expression of inflammatory mediators by oral epithelial cells that cause ARDS in COVID-19 patients. In addition, we report that MASL also inhibits SARS-CoV-2 infection of kidney epithelial cells in culture. This work identifies MASL as an agent with potential to inhibit SARS-CoV-2 infection and COVID-19 related inflammatory syndromes.


Asunto(s)
Antivirales/farmacología , Tratamiento Farmacológico de COVID-19 , Lectinas/farmacología , Boca/efectos de los fármacos , SARS-CoV-2/efectos de los fármacos , Glicoproteína de la Espiga del Coronavirus/efectos de los fármacos , Progresión de la Enfermedad , Células Epiteliales/efectos de los fármacos , Células Epiteliales/metabolismo , Interacciones Microbiota-Huesped/efectos de los fármacos , Humanos , Maackia/metabolismo , SARS-CoV-2/patogenicidad , Glicoproteína de la Espiga del Coronavirus/metabolismo
5.
Cancer Sci ; 109(5): 1292-1299, 2018 May.
Artículo en Inglés | MEDLINE | ID: mdl-29575529

RESUMEN

Podoplanin (PDPN) is a transmembrane receptor glycoprotein that is upregulated on transformed cells, cancer associated fibroblasts and inflammatory macrophages that contribute to cancer progression. In particular, PDPN increases tumor cell clonal capacity, epithelial mesenchymal transition, migration, invasion, metastasis and inflammation. Antibodies, CAR-T cells, biologics and synthetic compounds that target PDPN can inhibit cancer progression and septic inflammation in preclinical models. This review describes recent advances in how PDPN may be used as a biomarker and therapeutic target for many types of cancer, including glioma, squamous cell carcinoma, mesothelioma and melanoma.


Asunto(s)
Antineoplásicos/farmacología , Glicoproteínas de Membrana/genética , Neoplasias/genética , Regulación hacia Arriba , Antineoplásicos/uso terapéutico , Fibroblastos Asociados al Cáncer/metabolismo , Línea Celular Tumoral , Movimiento Celular , Progresión de la Enfermedad , Transición Epitelial-Mesenquimal/efectos de los fármacos , Regulación Neoplásica de la Expresión Génica/efectos de los fármacos , Humanos , Glicoproteínas de Membrana/metabolismo , Terapia Molecular Dirigida , Neoplasias/tratamiento farmacológico , Neoplasias/inmunología , Neoplasias/metabolismo , Regulación hacia Arriba/efectos de los fármacos
6.
Exp Cell Res ; 335(1): 115-22, 2015 Jul 01.
Artículo en Inglés | MEDLINE | ID: mdl-25959509

RESUMEN

Podoplanin (PDPN) is a transmembrane glycoprotein that promotes tumor cell migration, invasion, and cancer metastasis. In fact, PDPN expression is induced in many types of cancer. Thus, PDPN has emerged as a functionally relevant cancer biomarker and chemotherapeutic target. PDPN contains 2 intracellular serine residues that are conserved between species ranging from mouse to humans. Recent studies indicate that protein kinase A (PKA) can phosphorylate PDPN in order to inhibit cell migration. However, the number and identification of specific residues phosphorylated by PKA have not been defined. In addition, roles of other kinases that may phosphorylate PDPN to control cell migration have not been investigated. We report here that cyclin dependent kinase 5 (CDK5) can phosphorylate PDPN in addition to PKA. Moreover, results from this study indicate that PKA and CDK5 cooperate to phosphorylate PDPN on both intracellular serine residues to decrease cell motility. These results provide new insight into PDPN phosphorylation dynamics and the role of PDPN in cell motility. Understanding novel mechanisms of PDPN intracellular signaling could assist with designing novel targeted chemotherapeutic agents and procedures.


Asunto(s)
Movimiento Celular , Proteínas Quinasas Dependientes de AMP Cíclico/metabolismo , Quinasa 5 Dependiente de la Ciclina/metabolismo , Glicoproteínas de Membrana/metabolismo , Serina/metabolismo , Animales , Línea Celular Tumoral , Proliferación Celular , Glicoproteínas de Membrana/genética , Ratones , Fosforilación , Estructura Terciaria de Proteína , Serina/genética
7.
Carcinogenesis ; 36 Suppl 1: S2-18, 2015 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-26106139

RESUMEN

As part of the Halifax Project, this review brings attention to the potential effects of environmental chemicals on important molecular and cellular regulators of the cancer hallmark of evading growth suppression. Specifically, we review the mechanisms by which cancer cells escape the growth-inhibitory signals of p53, retinoblastoma protein, transforming growth factor-beta, gap junctions and contact inhibition. We discuss the effects of selected environmental chemicals on these mechanisms of growth inhibition and cross-reference the effects of these chemicals in other classical cancer hallmarks.


Asunto(s)
Exposición a Riesgos Ambientales/efectos adversos , Sustancias Peligrosas/efectos adversos , Neoplasias/inducido químicamente , Neoplasias/etiología , Animales , Humanos , Transducción de Señal/efectos de los fármacos
8.
Carcinogenesis ; 36 Suppl 1: S254-96, 2015 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-26106142

RESUMEN

Lifestyle factors are responsible for a considerable portion of cancer incidence worldwide, but credible estimates from the World Health Organization and the International Agency for Research on Cancer (IARC) suggest that the fraction of cancers attributable to toxic environmental exposures is between 7% and 19%. To explore the hypothesis that low-dose exposures to mixtures of chemicals in the environment may be combining to contribute to environmental carcinogenesis, we reviewed 11 hallmark phenotypes of cancer, multiple priority target sites for disruption in each area and prototypical chemical disruptors for all targets, this included dose-response characterizations, evidence of low-dose effects and cross-hallmark effects for all targets and chemicals. In total, 85 examples of chemicals were reviewed for actions on key pathways/mechanisms related to carcinogenesis. Only 15% (13/85) were found to have evidence of a dose-response threshold, whereas 59% (50/85) exerted low-dose effects. No dose-response information was found for the remaining 26% (22/85). Our analysis suggests that the cumulative effects of individual (non-carcinogenic) chemicals acting on different pathways, and a variety of related systems, organs, tissues and cells could plausibly conspire to produce carcinogenic synergies. Additional basic research on carcinogenesis and research focused on low-dose effects of chemical mixtures needs to be rigorously pursued before the merits of this hypothesis can be further advanced. However, the structure of the World Health Organization International Programme on Chemical Safety 'Mode of Action' framework should be revisited as it has inherent weaknesses that are not fully aligned with our current understanding of cancer biology.


Asunto(s)
Carcinogénesis/inducido químicamente , Carcinógenos Ambientales/efectos adversos , Exposición a Riesgos Ambientales/efectos adversos , Sustancias Peligrosas/efectos adversos , Neoplasias/inducido químicamente , Neoplasias/etiología , Animales , Humanos
9.
Ann Rheum Dis ; 74(1): 275-84, 2015 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-24225059

RESUMEN

OBJECTIVE: This study investigated whether chondrocytes within the cartilage matrix have the capacity to communicate through intercellular connections mediated by voltage-gated gap junction (GJ) channels. METHODS: Frozen cartilage samples were used for immunofluorescence and immunohistochemistry assays. Samples were embedded in cacodylate buffer before dehydration for scanning electron microscopy. Co-immunoprecipitation experiments and mass spectrometry (MS) were performed to identify proteins that interact with the C-terminal end of Cx43. GJ communication was studied through in situ electroporation, electrophysiology and dye injection experiments. A transwell layered culture system and MS were used to identify and quantify transferred amino acids. RESULTS: Microscopic images revealed the presence of multiple cellular projections connecting chondrocytes within the matrix. These projections were between 5 and 150 µm in length. MS data analysis indicated that the C-terminus of Cx43 interacts with several cytoskeletal proteins implicated in Cx trafficking and GJ assembly, including α-tubulin and ß-tubulin, actin, and vinculin. Electrophysiology experiments demonstrated that 12-mer oligonucleotides could be transferred between chondrocytes within 12 min after injection. Glucose was homogeneously distributed within 22 and 35 min. No transfer was detected when glucose was electroporated into A549 cells, which have no GJs. Transwell layered culture systems coupled with MS analysis revealed connexins can mediate the transfer of L-lysine and L-arginine between chondrocytes. CONCLUSIONS: This study reveals that intercellular connections between chondrocytes contain GJs that play a key role in cell-cell communication and a metabolic function by exchange of nutrients including glucose and essential amino acids. A three-dimensional cellular network mediated through GJs might mediate metabolic and physiological homeostasis to maintain cartilage tissue.


Asunto(s)
Cartílago Articular/metabolismo , Comunicación Celular , Condrocitos/metabolismo , Conexinas/metabolismo , Uniones Comunicantes/metabolismo , Aminoácidos Esenciales/metabolismo , Animales , Cartílago Articular/ultraestructura , Condrocitos/ultraestructura , Conexinas/ultraestructura , Uniones Comunicantes/ultraestructura , Glucosa/metabolismo , Homeostasis , Humanos , Inmunohistoquímica , Inmunoprecipitación , Articulación de la Rodilla , Microscopía Electrónica de Rastreo , Porcinos
10.
J Biol Chem ; 288(17): 12215-21, 2013 Apr 26.
Artículo en Inglés | MEDLINE | ID: mdl-23530051

RESUMEN

Podoplanin (PDPN) is a transmembrane receptor that affects the activities of Rho, ezrin, and other proteins to promote tumor cell motility, invasion, and metastasis. PDPN is found in many types of cancer and may serve as a tumor biomarker and chemotherapeutic target. The intracellular region of PDPN contains only two serines, and these are conserved in mammals including mice and humans. We generated cells from the embryos of homozygous null Pdpn knock-out mice to investigate the relevance of these serines to cell growth and migration on a clear (PDPN-free) background. We report here that one or both of these serines can be phosphorylated by PKA (protein kinase A). We also report that conversion of these serines to nonphosphorylatable alanine residues enhances cell migration, whereas their conversion to phosphomimetic aspartate residues decreases cell migration. These results indicate that PKA can phosphorylate PDPN to decrease cell migration. In addition, we report that PDPN expression in fibroblasts causes them to facilitate the motility and viability of neighboring melanoma cells in coculture. These findings shed new light on how PDPN promotes cell motility, its role in tumorigenesis, and its utility as a functionally relevant biomarker and chemotherapeutic target.


Asunto(s)
Movimiento Celular , Proteínas Quinasas Dependientes de AMP Cíclico/metabolismo , Fibroblastos/metabolismo , Melanoma/metabolismo , Glicoproteínas de Membrana/metabolismo , Proteínas de Neoplasias/metabolismo , Animales , Línea Celular Tumoral , Técnicas de Cocultivo , Proteínas Quinasas Dependientes de AMP Cíclico/genética , Fibroblastos/patología , Melanoma/genética , Melanoma/patología , Glicoproteínas de Membrana/genética , Ratones , Ratones Noqueados , Proteínas de Neoplasias/genética , Fosforilación/genética , Serina/genética , Serina/metabolismo
11.
J Med Educ Curric Dev ; 10: 23821205231177862, 2023.
Artículo en Inglés | MEDLINE | ID: mdl-37275656

RESUMEN

Problem-based learning (PBL) utilizes a self-directed strategy. This process relies on group participation to succeed. Students without a background in biology or medicine can feel overwhelmed by the complexity of the subject matter and unable to participate in the group learning process. We incorporated curated educational videos in the PBL curriculum to help address this situation. First year medical students participated in this study in the form of a typical PBL session. They were then assessed on basic and clinical science knowledge and their learning experience. Student basic science and clinical knowledge were similar between the student groups. However, the students given a list of suggested videos scored higher in their learning experience, perception of feeling prepared, and participating in the group PBL experience than students who were not given the video list. Results from this study indicate that videos can be utilized to enhance the PBL process.

12.
Biochem Biophys Rep ; 32: 101341, 2022 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-36120492

RESUMEN

Up to 70 million people around the world suffer from rheumatoid arthritis. Current treatment options have varied efficacy and can cause unwanted side effects. New approaches are needed to treat this condition. Sialic acid modifications on chondrocyte receptors have been associated with arthritic inflammation and joint destruction. For example, the transmembrane mucin receptor protein podoplanin (PDPN) has been identified as a functionally relevant receptor that presents extracellular sialic acid motifs. PDPN signaling promotes inflammation and invasion associated with arthritis and, therefore, has emerged as a target that can be used to inhibit arthritic inflammation. Maackia amurensis seed lectin (MASL) can target PDPN on chondrocytes to decrease inflammatory signaling cascades and reduce cartilage destruction in a lipopolysaccharide induced osteoarthritis mouse model. Here, we investigated the effects of MASL on rheumatoid arthritis progression in a TNFα transgenic (TNF-Tg) mouse model. Results from this study indicate that MASL can be administered orally to ameliorate joint malformation and increase velocity of movement exhibited by these TNF-Tg mice. These data support the consideration of MASL as a potential treatment for rheumatoid arthritis.

13.
J Biol Chem ; 285(13): 9649-9656, 2010 Mar 26.
Artículo en Inglés | MEDLINE | ID: mdl-20123990

RESUMEN

Nontransformed cells can force tumor cells to assume a normal morphology and phenotype by the process of contact normalization. Transformed cells must escape this process to become invasive and malignant. However, mechanisms underlying contact normalization have not been elucidated. Here, we have identified genes that are affected by contact normalization of Src-transformed cells. Tumor cells must migrate to become invasive and malignant. Src must phosphorylate the adaptor protein Cas (Crk-associated substrate) to promote tumor cell motility. We report here that Src utilizes Cas to induce podoplanin (Pdpn) expression to promote tumor cell migration. Pdpn is a membrane-bound extracellular glycoprotein that associates with endogenous ligands to promote tumor cell migration leading to cancer invasion and metastasis. In fact, Pdpn expression accounted for a major part of the increased migration seen in Src-transformed cells. Moreover, nontransformed cells suppressed Pdpn expression in adjacent Src-transformed cells. Of >39,000 genes, Pdpn was one of only 23 genes found to be induced by transforming Src activity and suppressed by contact normalization of Src-transformed cells. In addition, we found 16 genes suppressed by Src and induced by contact normalization. These genes encode growth factor receptors, adaptor proteins, and products that have not yet been annotated and may play important roles in tumor cell growth and migration.


Asunto(s)
Proteína Sustrato Asociada a CrK/metabolismo , Regulación Enzimológica de la Expresión Génica , Regulación Neoplásica de la Expresión Génica , Glicoproteínas de Membrana/biosíntesis , Familia-src Quinasas/metabolismo , Animales , Línea Celular Transformada , Movimiento Celular , Fibroblastos/metabolismo , Homocigoto , Ligandos , Ratones , Microscopía Fluorescente/métodos , Modelos Biológicos , Análisis de Secuencia por Matrices de Oligonucleótidos , ARN Interferente Pequeño/metabolismo
14.
J Am Coll Nutr ; 30(4): 233-40, 2011 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-21917703

RESUMEN

OBJECTIVE: To examine the association of high-sensitivity C-reactive protein (hsCRP), a systemic biomarker for the inflammatory process at entry to care, with pregnancy-induced hypertension/preeclampsia, adverse outcomes of pregnancy, and the maternal diet. DESIGN: Random sample (N = 520) with normal glucose tolerance from a large prospective cohort study of urban, low income, minority gravidae. RESULTS: During pregnancy, the highest tertile of hsCRP (range, 7.06-137.41 mg/L) was associated with significantly increased risks for early preterm delivery (<34 weeks). However, after stratification by maternal pregravid body mass index (BMI), risk for early preterm delivery <34 weeks (adjusted odds ratios [AOR] = 3.58, 95% confidence interval [CI] = 1.05-12.27), and pregnancy-induced hypertension (AOR = 2.66, 95% CI = 1.03-6.86) including preeclampsia (AOR = 2.72, 95% CI = 1.08-6.85) was shown to be specific to lean women (BMI <25) with high hsCRP. Increased hsCRP was unrelated to risk among overweight and obese gravidae. We found high hsCRP to be associated with diet. After stratification by BMI, dietary differences (higher intakes of protein and cholesterol with a lower intake of carbohydrate and a higher entry dietary glycemic index) were associated with increased hsCRP only among lean gravidae and not among those who were overweight or obese. CONCLUSIONS: High hsCRP is a diet-related biomarker for serious complications and poor outcome in lean women with normal glucose tolerance.


Asunto(s)
Proteína C-Reactiva/análisis , Fenómenos Fisiologicos Nutricionales Maternos , Resultado del Embarazo , Adulto , Biomarcadores/análisis , Dieta , Femenino , Número de Embarazos , Humanos , Modelos Logísticos , Obesidad/complicaciones , Oportunidad Relativa , Preeclampsia/etiología , Embarazo , Nacimiento Prematuro/etiología , Estudios Prospectivos , Factores de Riesgo , Encuestas y Cuestionarios , Adulto Joven
15.
J Cancer Res Clin Oncol ; 147(2): 445-457, 2021 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-33205348

RESUMEN

PURPOSE: Oral cancer causes over 120,000 deaths annually and affects the quality of life for survivors. Over 90% of oral cancers are derived from oral squamous cell carcinoma cells (OSCCs) which are generally resistant to standard cytotoxic chemotherapy agents. OSCC cells often exhibit increased TGFß and PDPN receptor activity compared to nontransformed oral epithelial cells. Maackia amurensis seed lectin (MASL) can target the PDPN receptor and has been identified as a novel agent that can be used to treat oral cancer. However, mechanisms by which MASL inhibits OSCC progression are not yet clearly defined. METHODS: Here, we performed cell migration and cytotoxicity assays to assess the effects of MASL on OSCC motility and viability at physiologically relevant concentrations. We then performed comprehensive transcriptome analysis combined with transcription factor reporter assays to investigate the how MASL affects OSCC gene expression at these concentration. Key data were then confirmed by western blotting to evaluate the effects of MASL on gene expression and kinase signaling activity at the protein level. RESULTS: MASL significantly affected the expression of about 27% of approximately 15,000 genes found to be expressed by HSC-2 cells used to model OSCC cells in this study. These genes affected by MASL include members of the TGFß-SMAD, JAK-STAT, and Wnt-ßCTN signaling pathways. In particular, MASL decreased expression of PDPN, SOX2, and SMAD5 at the RNA and protein levels. MASL also inhibited SMAD and MAPK activity, and exhibited potential for combination therapy with doxorubicin and 5-fluorouracil. CONCLUSIONS: Taken together, results from this study indicate that MASL decreases activity of JAK-STAT, TGFß-SMAD, and Wnt-ßCTN signaling pathways to inhibit OSCC growth and motility. These data suggest that further studies should be undertaken to determine how MASL may also be used alone and in combination with other agents to treat oral cancer.


Asunto(s)
Regulación Neoplásica de la Expresión Génica/efectos de los fármacos , Maackia/química , Neoplasias de la Boca/tratamiento farmacológico , Lectinas de Plantas/farmacología , Carcinoma de Células Escamosas de Cabeza y Cuello/tratamiento farmacológico , Movimiento Celular/efectos de los fármacos , Humanos , Neoplasias de la Boca/metabolismo , Neoplasias de la Boca/patología , Lectinas de Plantas/uso terapéutico , Factores de Transcripción SOXB1/genética , Transducción de Señal/efectos de los fármacos , Proteínas Smad/genética , Carcinoma de Células Escamosas de Cabeza y Cuello/metabolismo , Carcinoma de Células Escamosas de Cabeza y Cuello/patología , Transcripción Genética/efectos de los fármacos , Vía de Señalización Wnt/efectos de los fármacos
16.
Res Sq ; 2020 Oct 23.
Artículo en Inglés | MEDLINE | ID: mdl-33106801

RESUMEN

COVID-19 was declared an international public health emergency in January, and a pandemic in March of 2020. There are over 23 million confirmed COVID-19 cases that have cause over 800 thousand deaths worldwide as of August 19th, 2020. COVID-19 is caused by the SARS-CoV-2 virus. SARS-CoV-2 presents a surface "spike" protein that binds to the ACE2 receptor to infect host cells. In addition to the respiratory tract, SARS-Cov-2 can also infect cells of the oral mucosa, which also express the ACE2 receptor. The spike and ACE2 proteins are highly glycosylated with sialic acid modifications that direct viral-host interactions and infection. Maackia amurensis seed lectin (MASL) has a strong affinity for sialic acid modified proteins and can be used as an antiviral agent. Here, we report that MASL targets the ACE2 receptor, decreases ACE2 expression and glycosylation, suppresses binding of the SARS-CoV-2 spike protein, and decreases expression of inflammatory mediators by oral epithelial cells that cause ARDS in COVID-19 patients. This work identifies MASL as an agent with potential to inhibit SARS-CoV-2 infection and COVID-19 related inflammatory syndromes.

17.
Sci Rep ; 10(1): 21959, 2020 12 15.
Artículo en Inglés | MEDLINE | ID: mdl-33319820

RESUMEN

Breast cancer is the leading cause of cancer death among women worldwide. Like other cancers, mammary carcinoma progression involves acidification of the tumor microenvironment, which is an important factor for cancer detection and treatment strategies. However, the effects of acidity on mammary carcinoma cell morphology and phenotype have not been thoroughly characterized. Here, we evaluated fundamental effects of environmental acidification on mammary carcinoma cells in standard two-dimensional cultures and three-dimensional spheroids. Acidification decreased overall mammary carcinoma cell viability, while increasing their resistance to the anthracycline doxorubicin. Environmental acidification also increased extracellular vesicle production by mammary carcinoma cells. Conditioned media containing these vesicles appeared to increase fibroblast motility. Acidification also increased mammary carcinoma cell motility when cultured with fibroblasts in spheroids. Taken together, results from this study suggest that environmental acidification induces drug resistance and extracellular vesicle production by mammary carcinoma cells that promote tumor expansion.


Asunto(s)
Ácidos/química , Concentración de Iones de Hidrógeno , Neoplasias Mamarias Animales/patología , Esferoides Celulares/metabolismo , Animales , Línea Celular Tumoral , Supervivencia Celular , Femenino , Humanos , Técnicas In Vitro , Neoplasias Mamarias Animales/metabolismo , Microambiente Tumoral
18.
Drug Discov Today ; 24(1): 241-249, 2019 01.
Artículo en Inglés | MEDLINE | ID: mdl-30077780

RESUMEN

Cancer and arthritis present an enormous challenge to society. They share pathogenic pathways that involve extracellular matrix degradation, tissue invasion, and inflammation. Most cancer and arthritis treatments affect normal cell function to cause significant adverse effects in patients. Specific pathways that promote cancer and arthritis progression must be elucidated to design more targeted and effective therapeutics. The Src kinase and podoplanin (PDPN) receptor are upregulated in cancer cells, fibroblasts, synoviocytes, and immune cells that increase tissue invasion and inflammation to promote both cancer and arthritis. In this review, we discuss how Src and PDPN forge a path to tissue destruction, and how they can serve as targets for therapeutics to combat cancer and arthritis.


Asunto(s)
Artritis/metabolismo , Glicoproteínas de Membrana/metabolismo , Neoplasias/metabolismo , Familia-src Quinasas/metabolismo , Animales , Humanos
19.
Cancer Sci ; 99(7): 1326-33, 2008 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-18422756

RESUMEN

The Src tyrosine kinase associates with the focal adhesion adaptor protein Cas (Crk-associated substrate) to suppress the expression of potential tumor suppressor genes. For example, Src utilizes Cas to suppress the expression of the LIM-only protein Fhl1 (four and a half LIM domains 1), in order to promote non-anchored tumor-cell growth and migration. Here, we report that the promoter region of the Fhl1 gene was methylated more in Src-transformed cells than non-transformed cells. In addition, global expression analysis indicates that Fhl1 induced expression of serum deprivation response factor (Sdpr) in Src-transformed cells. Moreover, Fhl1 and Sdpr was expressed in approximately 87% and 40% of samples obtained from non-transformed breast, 100% of samples obtained from non-transformed kidney, and over 60% of samples obtained from non-transformed prostate. In contrast, Fhl1 and Sdpr was detected in approximately 40% and 7% of matched samples from mammary carcinoma, less than 11% of matched samples from kidney carcinoma, and in less than 22% of matched samples from prostate carcinoma. These data indicate that Fhl1 and Sdpr expression was significantly reduced in tumors of the breast (P < 0.02 and P < 0.001), kidney (P < 0.01), and prostate (P < 0.05). In addition, although Src can activate mitogen-activated protein kinase (MAPK) to promote tumor-cell growth, our data indicate that Src did not rely on MAPK activity to suppress the expression of Fhl1 and Sdpr in transformed cells. Thus, Src induced methylation of the promoter region of the Fhl1 gene; Src suppressed Fhl1 and Sdpr expression independent of mitogen-activated protein kinase (MAPK) activity; Fhl1 induced the expression of Sdpr in Src-transformed cells; and Fhl1 and Sdpr expression was suppressed in tumors of the breast, kidney, and prostate.


Asunto(s)
Neoplasias de la Mama/química , Proteínas Portadoras/análisis , Péptidos y Proteínas de Señalización Intracelular/análisis , Neoplasias Renales/química , Proteínas Musculares/análisis , Neoplasias de la Próstata/química , Animales , Proteínas Portadoras/genética , Transformación Celular Neoplásica , Proteína Sustrato Asociada a CrK/fisiología , Metilación de ADN , Progresión de la Enfermedad , Femenino , Humanos , Inmunohistoquímica , Péptidos y Proteínas de Señalización Intracelular/genética , Proteínas con Dominio LIM , Masculino , Ratones , Proteínas Musculares/genética , Células 3T3 NIH , Proteínas de Unión a Fosfato , Regiones Promotoras Genéticas , Familia-src Quinasas/fisiología
20.
Cancer Res ; 66(3): 1543-52, 2006 Feb 01.
Artículo en Inglés | MEDLINE | ID: mdl-16452211

RESUMEN

Anchorage independence and motility are hallmarks of tumor cell growth. Tumor cell growth and morphology can be normalized by contact with nontransformed cells. The Src tyrosine kinase phosphorylates specific sites on the focal adhesion adaptor protein Crk-associated substrate (Cas) to promote nonanchored cell growth and migration. We studied the effects of Src and Cas on the expression of >14,000 genes to identify molecular events that underlie these activities. Gene expression in tumor cells that were normalized by neighboring nontransformed cells was used as an additional filter to identify genes that control metastatic cell growth. This process enabled the identification of genes that play roles in anchorage-independent cell growth and migration. One candidate, four and a half LIM domains 1 (Fhl1), acts as a transcriptional regulator that can associate with cell junctions as well as with the nucleus. We show here that Src phosphorylates Cas to block Fhl1 expression. In addition, suppression of Fhl1 is required for Src to promote tumor cell growth. These data show that Fhl1 is a tumor suppressor gene that acts downstream of Src and Cas to specifically block anchorage-independent cell growth and migration. Moreover, Fhl1 was suppressed in tumors from several human tissues. Thus, identification of how Fhl1 controls fundamental aspects of tumor cell growth and metastasis may lead to the development of novel markers that can be used to diagnose human clinical specimens as well as open innovative avenues of investigations aimed at developing reagents that target cancer cells while minimizing damage to normal cells.


Asunto(s)
Movimiento Celular/fisiología , Proteína de Susceptibilidad a Apoptosis Celular/metabolismo , Proteínas Musculares/antagonistas & inhibidores , Neoplasias Experimentales/patología , Familia-src Quinasas/metabolismo , Secuencia de Aminoácidos , Animales , Adhesión Celular/genética , Adhesión Celular/fisiología , Procesos de Crecimiento Celular/genética , Procesos de Crecimiento Celular/fisiología , Línea Celular Tumoral , Movimiento Celular/genética , Regulación Neoplásica de la Expresión Génica , Genes Supresores de Tumor , Humanos , Péptidos y Proteínas de Señalización Intracelular , Proteínas con Dominio LIM , Ratones , Ratones Noqueados , Datos de Secuencia Molecular , Proteínas Musculares/biosíntesis , Proteínas Musculares/genética , Neoplasias Experimentales/genética , Neoplasias Experimentales/metabolismo , Fosforilación
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA