Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 16 de 16
Filter
1.
Int J Mol Sci ; 25(4)2024 Feb 06.
Article in English | MEDLINE | ID: mdl-38396665

ABSTRACT

Mesenchymal stem/stromal cells (MSCs) are multipotent cells located in different areas of the human body. The oral cavity is considered a potential source of MSCs because they have been identified in several dental tissues (D-MSCs). Clinical trials in which cells from these sources were used have shown that they are effective and safe as treatments for tissue regeneration. Importantly, immunoregulatory capacity has been observed in all of these populations; however, this function may vary among the different types of MSCs. Since this property is of clinical interest for cell therapy protocols, it is relevant to analyze the differences in immunoregulatory capacity, as well as the mechanisms used by each type of MSC. Interestingly, D-MSCs are the most suitable source for regenerating mineralized tissues in the oral region. Furthermore, the clinical potential of D-MSCs is supported due to their adequate capacity for proliferation, migration, and differentiation. There is also evidence for their potential application in protocols against autoimmune diseases and other inflammatory conditions due to their immunosuppressive capacity. Therefore, in this review, the immunoregulatory mechanisms identified at the preclinical level in combination with the different types of MSCs found in dental tissues are described, in addition to a description of the clinical trials in which MSCs from these sources have been applied.


Subject(s)
Mesenchymal Stem Cells , Humans , Mesenchymal Stem Cells/metabolism , Immunomodulation , Multipotent Stem Cells , Cell Differentiation , Cell- and Tissue-Based Therapy , Cell Proliferation , Cells, Cultured
2.
Cells ; 12(7)2023 03 30.
Article in English | MEDLINE | ID: mdl-37048119

ABSTRACT

Macrophages with the M2 phenotype promote tumor development through the immunosuppression of antitumor immunity. We previously demonstrated the presence of mesenchymal stem/stromal cells (MSCs) in cervical cancer (CeCa-MSCs), suggesting an immune protective capacity in tumors, but to date, their effect in modulating macrophage polarization remains unknown. In this study, we compared the capacities of MSCs from normal cervix (NCx) and CeCa to promote M2 macrophage polarization in a coculture system. Our results demonstrated that CeCa-MSCs, in contrast to NCx-MSCs, significantly decreased M1 macrophage cell surface marker expression (HLA-DR, CD80, CD86) and increased M2 macrophage expression (CD14, CD163, CD206, Arg1) in cytokine-induced CD14+ monocytes toward M1- or M2-polarized macrophages. Interestingly, compared with NCx-MSCs, in M2 macrophages generated from CeCa-MSC cocultures, we observed an increase in the percentage of phagocytic cells, in the intracellular production of IL-10 and IDO, the capacity to decrease T cell proliferation and for the generation of CD4+CD25+FoxP3+ Tregs. Importantly, this capacity to promote M2 macrophage polarization was correlated with the intracellular expression of macrophage colony-stimulating factor (M-CSF) and upregulation of IL-10 in CeCa-MSCs. Furthermore, the presence of M2 macrophages was correlated with the increased production of IL-10 and IL-1RA anti-inflammatory molecules. Our in vitro results indicate that CeCa-MSCs, in contrast to NCx-MSCs, display an increased M2-macrophage polarization potential and suggest a role of CeCa-MSCs in antitumor immunity.


Subject(s)
Interleukin-10 , Uterine Cervical Neoplasms , Humans , Female , Interleukin-10/metabolism , Uterine Cervical Neoplasms/metabolism , Macrophages/metabolism , Cytokines/metabolism , Stromal Cells/metabolism
3.
Reprod Toxicol ; 110: 150-160, 2022 06.
Article in English | MEDLINE | ID: mdl-35460820

ABSTRACT

Amphetamine derivatives negatively impact serotonin (5-HT) production, which triggers apoptosis in different tissues, depending on the receptor they bind. 5-HT in the ovary stimulates estradiol secretion, a survival factor of granulosa cells. The effect of amphetamine derivatives on the serotonergic system of the ovary and follicular development is unknown. Therefore, in this study, we investigated the effects of p-chloroamphetamine (pCA), derived from amphetamines, on estradiol production, follicular development, apoptosis of granulosa cells, and serotonin 5-HT7 receptor (R5-HT7) expression. Female rats (30 days old) were injected with 10 mg/kg of pCA intraperitoneally and were euthanized 48 or 120 h after treatment. The concentration of 5-HT in the hypothalamus decreased at 48 and 120 h after treatment and in the ovary at 120 h. The serum concentration of estradiol decreased at all times studied. Follicular atresia, TUNEL-positive (apoptotic) granulosa cells and Bax expression were elevated by pCA, but none of these effects was associated with R5-HT7 expression. These results suggest that pCA induces the dysregulation of the serotonergic system in the hypothalamus and the ovary, negatively impacting estradiol production and follicular development.


Subject(s)
Follicular Atresia , Serotonin , Amphetamine , Animals , Apoptosis , Estradiol/metabolism , Female , Follicular Atresia/physiology , Granulosa Cells/metabolism , Rats , p-Chloroamphetamine/pharmacology
4.
Int J Mol Sci ; 22(17)2021 Sep 02.
Article in English | MEDLINE | ID: mdl-34502453

ABSTRACT

Mesenchymal stem/stromal cells (MSCs) have an immunoregulatory capacity and have been used in different clinical protocols requiring control of the immune response. However, variable results have been obtained, mainly due to the effect of the microenvironment on the induction, increase, and maintenance of MSC immunoregulatory mechanisms. In addition, the importance of cell-cell contact for MSCs to efficiently modulate the immune response has recently been highlighted. Because these interactions would be difficult to achieve in the physiological context, the release of extracellular vesicles (EVs) and their participation as intermediaries of communication between MSCs and immune cells becomes relevant. Therefore, this article focuses on analyzing immunoregulatory mechanisms mediated by cell contact, highlighting the importance of intercellular adhesion molecule-1 (ICAM-1) and the participation of EVs. Moreover, the effects of tumor necrosis factor-alpha (TNF-α) and interferon-gamma (IFN-γ), the main cytokines involved in MSC activation, are examined. These cytokines, when used at the appropriate concentrations and times, would promote increases in the expression of immunoregulatory molecules in the cell and allow the acquisition of EVs enriched with these molecules. The establishment of certain in vitro activation guidelines will facilitate the design of conditioning protocols to obtain functional MSCs or EVs in different pathophysiological conditions.


Subject(s)
Cell Communication , Immunomodulation , Interferon-gamma/physiology , Mesenchymal Stem Cells/immunology , Tumor Necrosis Factor-alpha/physiology , Animals , Cytokines/metabolism , Extracellular Vesicles/metabolism , Humans
5.
J Immunol Res ; 2020: 8839625, 2020.
Article in English | MEDLINE | ID: mdl-33335929

ABSTRACT

Bone marrow mesenchymal stem/stromal cells (BM-MSCs) have immunoregulatory capacity; therefore, they have been used in different clinical protocols in which it is necessary to decrease the immune response. This capacity is mainly regulated by TNF-α and IFN-γ, and it has been observed that cell-cell contact, mainly mediated by ICAM-1, is important for MSCs to carry out efficient immunoregulation. Therefore, in the present work, we analyzed the effect of TNF-α alone or in combination with IFN-γ on the expression of ICAM-1. Besides, given the importance of cell contact in the immunoregulatory function of MSCs, we analyzed whether these cells release ICAM-1+ microvesicles (MVs). Our results show for the first time that TNF-α is capable of increasing the early expression of ICAM-1 in human BM-MSCs. Also, we observed that TNF-α and IFN-γ have a synergistic effect on the increase in the expression of ICAM-1. Furthermore, we found that BM-MSCs exposed to an inflammatory environment release MVs enriched in ICAM-1 (MVs-ICAM-1high). The knowledge generated in this study will contribute to the improvement of in vitro conditioning protocols that favor the therapeutic effect of these cells or their products.


Subject(s)
Cell-Derived Microparticles/metabolism , Cellular Microenvironment , Inflammation/metabolism , Intercellular Adhesion Molecule-1/genetics , Mesenchymal Stem Cells/metabolism , Biomarkers , Cell Adhesion Molecules/genetics , Cell Adhesion Molecules/metabolism , Cytokines/metabolism , Disease Susceptibility , Histocompatibility Antigens Class I/genetics , Histocompatibility Antigens Class I/immunology , Humans , Immunophenotyping , Inflammation/etiology , Inflammation/pathology , Inflammation Mediators/metabolism , Interferon-gamma/metabolism , Leukocytes, Mononuclear/immunology , Leukocytes, Mononuclear/metabolism , Models, Biological , Tumor Necrosis Factor-alpha/metabolism
6.
Stem Cells Int ; 2017: 6061729, 2017.
Article in English | MEDLINE | ID: mdl-29675046

ABSTRACT

Mesenchymal stem/stromal cells (MSCs) from bone marrow (BM) have been used in coculture systems as a feeder layer for promoting the expansion of hematopoietic progenitor cells (HPCs) for hematopoietic cell transplantation. Because BM has some drawbacks, umbilical cord blood (UCB) and placenta (PL) have been proposed as possible alternative sources of MSCs. However, MSCs from UCB and PL sources have not been compared to determine which of these cell populations has the best capacity of promoting hematopoietic expansion. In this study, MSCs from UCB and PL were cultured under the same conditions to compare their capacities to support the expansion of HPCs in vitro. MSCs were cocultured with CD34+CD38-Lin- HPCs in the presence or absence of early acting cytokines. HPC expansion was analyzed through quantification of colony-forming cells (CFCs), long-term culture-initiating cells (LTC-ICs), and CD34+CD38-Lin- cells. MSCs from UCB and PL have similar capacities to increase HPC expansion, and this capacity is similar to that presented by BM-MSCs. Here, we are the first to determine that MSCs from UCB and PL have similar capacities to promote HPC expansion; however, PL is a better alternative source because MSCs can be obtained from a higher proportion of samples.

7.
Neurochem Res ; 42(2): 415-427, 2017 Feb.
Article in English | MEDLINE | ID: mdl-27804011

ABSTRACT

Neural progenitors (NP), found in fetal and adult brain, differentiate into neurons potentially able to be used in cell replacement therapies. This approach however, raises technical and ethical problems which limit their potential therapeutic use. Alternately, NPs can be obtained by transdifferentiation of non-neural somatic cells evading these difficulties. Human bone marrow mesenchymal stromal cells (MSCs) are suggested to transdifferentiate into NP-like cells, which however, have a low proliferation capacity. The present study demonstrates the requisite of cell adhesion for proliferation and survival of NP-like cells and re-evaluates some neuronal features after differentiation by standard procedures. Mature neuronal markers, though, were not detected by these procedures. A chemical differentiation approach was used in this study to convert MSCs-derived NP-like cells into neurons by using a cocktail of six molecules, CHIR99021, I-BET151, RepSox, DbcAMP, forskolin and Y-27632, defined after screening combinations of 22 small molecules. Direct transdifferentiation of MSCs into neuronal cells was obtained with the small molecule cocktail, without requiring the NP-like intermediate stage.


Subject(s)
Cell Proliferation/physiology , Cell Transdifferentiation/physiology , Mesenchymal Stem Cells/physiology , Neural Stem Cells/physiology , Neurons/physiology , Adolescent , Adult , Amides/administration & dosage , Cell Proliferation/drug effects , Cell Transdifferentiation/drug effects , Cells, Cultured , Colforsin/administration & dosage , Drug Combinations , Heterocyclic Compounds, 4 or More Rings/administration & dosage , Humans , Male , Mesenchymal Stem Cells/drug effects , Neural Stem Cells/drug effects , Neurons/drug effects , Pyridines/administration & dosage , Young Adult
8.
Bol. méd. Hosp. Infant. Méx ; 73(6): 380-387, Nov.-Dec. 2016. graf
Article in English | LILACS | ID: biblio-951256

ABSTRACT

Abstract: The relevance of the microenvironment in the initiation, promotion, and progression of cancer has been postulated. Mesenchymal stem cells (MSCs) have been identified as important components of the tumor stroma, which are capable of affecting the development of cancer through various mechanisms. In particular, MSCs immunosuppressive properties play an important role. It has been shown that bone marrow-derived and other healthy tissues-derived MSCs are capable of regulating the immune response by affecting the activation, maturation, proliferation, differentiation, and effector function of cells of the immune system, such as neutrophils, macrophages, dendritic cells, natural killer cells (NK) and T-lymphocytes. Similar mechanisms have been identified in MSCs associated with different types of tumors, where they generate an immunosuppressive microenvironment by decreasing the cytotoxic activity of T-lymphocytes and NK cells, skew macrophage differentiation towards an M2 phenotype, and decrease the secretion of Th1-type cytokines. Also, the cytokines, chemokines, and factors secreted by the transformed cells or other cells from the tumor stroma are capable of modulating the functions of MSCs.


Resumen: Se ha postulado la relevancia del microambiente en la iniciación, promoción y progresión del cáncer. Las células troncales mesenquimales (MSC, del inglés mesenchymal stem cells) se han identificado como un componente fundamental del estroma tumoral. Estas son capaces de favorecer el desarrollo del cáncer mediante varios mecanismos. En particular, sus propiedades inmunosupresoras juegan un papel importante. Se ha demostrado que las MSC de médula ósea y otros tejidos sanos son capaces de regular la respuesta inmune al afectar la activación, maduración, proliferación, diferenciación y función efectora de las células del sistema inmune, como neutrófilos, macrófagos, células dendríticas, células NK y linfocitos T. Mecanismos similares se han identificado en las MSC asociadas con diferentes tipos de tumores, donde estas se encargan de generar un microambiente inmunosupresor al disminuir la actividad citotóxica de linfocitos T y células NK, polarizar a los macrófagos hacia un fenotipo M2, y disminuir el patrón de secreción de citocinas tipo Th1. Asimismo, las citocinas, quimiocinas y factores secretados por las células transformadas u otras células del estroma tumoral son capaces de modular las funciones de las MSC.

9.
Stem Cells Dev ; 25(17): 1299-310, 2016 09 01.
Article in English | MEDLINE | ID: mdl-27462977

ABSTRACT

The differentiation capacity, hematopoietic support, and immunomodulatory properties of human bone marrow mesenchymal stromal cells (BM-MSCs) make them attractive therapeutic agents for a wide range of diseases. Clinical scale cultures (CSCs) have been used to expand BM-MSCs for their use in cell therapy protocols; however, little is known about the functionality of the expanded cells. The main goal of the present study was to evaluate the functional characteristics of BM-MSCs expanded from CSCs to determine the quality of the cells for cellular therapy protocols. To address this issue, we analyzed the morphology, immunophenotype, differentiation potential (adipogenic, osteogenic and chondrogenic), hematopoietic support, and immunosuppressive capacity of BM-MSCs from short scale cultures (SSCs) and CSCs in a comparative manner. After 12 days of culture in CSCs (HYPERFlask System), BM-MSCs reached cell numbers of 125.52 × 10(6) ± 25.6 × 10(6) MSCs, which corresponded to the number of cells required for transplantation (∼1.7 × 10(6) MSCs/kg for a 70-kg patient). After expansion, BM-MSCs expressed the characteristic markers CD73, CD90, and CD105; however, expansion decreased their differentiation capacity toward the adipogenic, osteogenic, and chondrogenic lineages and their ability to inhibit T-cell proliferation compared with SSCs-MSCs. Importantly, CSCs-MSCs maintained the ability to support the proliferation and expansion of hematopoietic progenitor cells and the capacity to express the molecules, cytokines, and extracellular matrix proteins involved in the regulation of hematopoiesis. Our study highlights the need to evaluate the functional properties of the expanded BM-MSCs for verification of their quality for cell therapy protocols.


Subject(s)
Bone Marrow Cells/cytology , Cell Culture Techniques/methods , Cell Differentiation , Hematopoietic Stem Cells/cytology , Immunosuppression Therapy , Mesenchymal Stem Cells/cytology , Adipogenesis/genetics , Antigens, CD/metabolism , Bone Marrow Cells/metabolism , Cell Differentiation/genetics , Cell Proliferation/genetics , Cell Shape/genetics , Cells, Cultured , Chondrogenesis/genetics , Cytokines/metabolism , Extracellular Matrix/metabolism , Gene Expression Regulation , Hematopoiesis/genetics , Hematopoietic Stem Cells/metabolism , Humans , Mesenchymal Stem Cells/metabolism
10.
Bol Med Hosp Infant Mex ; 73(6): 380-387, 2016.
Article in English | MEDLINE | ID: mdl-29421282

ABSTRACT

The relevance of the microenvironment in the initiation, promotion, and progression of cancer has been postulated. Mesenchymal stem cells (MSCs) have been identified as important components of the tumor stroma, which are capable of affecting the development of cancer through various mechanisms. In particular, MSCs immunosuppressive properties play an important role. It has been shown that bone marrow-derived and other healthy tissues-derived MSCs are capable of regulating the immune response by affecting the activation, maturation, proliferation, differentiation, and effector function of cells of the immune system, such as neutrophils, macrophages, dendritic cells, natural killer cells (NK) and T-lymphocytes. Similar mechanisms have been identified in MSCs associated with different types of tumors, where they generate an immunosuppressive microenvironment by decreasing the cytotoxic activity of T-lymphocytes and NK cells, skew macrophage differentiation towards an M2 phenotype, and decrease the secretion of Th1-type cytokines. Also, the cytokines, chemokines, and factors secreted by the transformed cells or other cells from the tumor stroma are capable of modulating the functions of MSCs.

11.
J Immunol Res ; 2015: 394917, 2015.
Article in English | MEDLINE | ID: mdl-25961059

ABSTRACT

Mesenchymal stem cells (MSCs) are multipotent cells capable of differentiation into mesenchymal lineages and that can be isolated from various tissues and easily cultivated in vitro. Currently, MSCs are of considerable interest because of the biological characteristics that confer high potential applicability in the clinical treatment of many diseases. Specifically, because of their high immunoregulatory capacity, MSCs are used as tools in cellular therapies for clinical protocols involving immune system alterations. In this review, we discuss the current knowledge about the capacity of MSCs for the immunoregulation of immunocompetent cells and emphasize the effects of MSCs on T cells, principal effectors of the immune response, and the immunosuppressive effects mediated by the secretion of soluble factors and membrane molecules. We also describe the mechanisms of MSC immunoregulatory modulation and the participation of MSCs as immune response regulators in several autoimmune diseases, and we emphasize the clinical application in graft versus host disease (GVHD).


Subject(s)
Autoimmune Diseases/immunology , Graft vs Host Disease/immunology , Hematopoiesis/immunology , Mesenchymal Stem Cells/immunology , B-Lymphocytes/cytology , B-Lymphocytes/immunology , Cell- and Tissue-Based Therapy , Dendritic Cells/cytology , Dendritic Cells/immunology , Humans , Immunomodulation , Killer Cells, Natural/cytology , Killer Cells, Natural/immunology , Mesenchymal Stem Cell Transplantation , T-Lymphocytes/cytology , T-Lymphocytes/immunology
12.
Stem Cells Dev ; 23(11): 1217-32, 2014 Jun 01.
Article in English | MEDLINE | ID: mdl-24428376

ABSTRACT

Bone marrow-mesenchymal stromal cells (BM-MSCs) have immunosuppressive properties and have been used in cell therapies as immune regulators for the treatment of graft-versus-host disease. We have previously characterized several biological properties of MSCs from placenta (PL) and umbilical cord blood (UCB), and compared them to those of BM-the gold standard. In the present study, we have compared MSCs from BM, UCB, and PL in terms of their immunosuppressive properties against lymphoid cell populations enriched for CD3(+) T cells. Our results confirm the immunosuppressive potential of BM-MSCs, and demonstrate that MSCs from UCB and, to a lesser extent PL, also have immunosuppressive potential. In contrast to PL-MSCs, BM-MSCs and UCB-MSCs significantly inhibited the proliferation of both CD4(+) and CD8(+) activated T cells in a cell-cell contact-dependent manner. Such a reduced proliferation in cell cocultures correlated with upregulation of programmed death ligand 1 on MSCs and cytotoxic T lymphocyte-associated Ag-4 (CTLA-4) on T cells, and increased production of interferon-γ, interleukin-10, and prostaglandin E2. Importantly, and in contrast to PL-MSCs, both BM-MSCs and UCB-MSCs favored the generation of T-cell subsets displaying a regulatory phenotype CD4(+)CD25(+)CTLA-4(+). Our results indicate that, besides BM-MSCs, UCB-MSCs might be a potent and reliable candidate for future therapeutic applications.


Subject(s)
Aging/physiology , Immune Tolerance/physiology , Mesenchymal Stem Cells/cytology , Mesenchymal Stem Cells/immunology , Primary Cell Culture , T-Lymphocytes/immunology , Adult , Adult Stem Cells/cytology , Adult Stem Cells/immunology , Cell Separation , Cells, Cultured , Coculture Techniques , Humans , Infant, Newborn , Lymphocyte Activation
13.
Stem Cells Dev ; 22(18): 2508-19, 2013 Sep 15.
Article in English | MEDLINE | ID: mdl-23656504

ABSTRACT

Mesenchymal stromal cells (MSCs) have been isolated from different tumors and it has been suggested that they support tumor growth through immunosuppression processes that favor tumor cell evasion from the immune system. To date, however, the presence of MSCs in cervical cancer (CeCa) and their possible role in tumor growth remains unknown. Herein we report on the presence of MSCs in cervical tissue, both in normal conditions (NCx-MSCs) and in CeCa (CeCa-MSCs), and described several biological properties of such cells. Our study showed similar patterns of cell surface antigen expression, but distinct differentiation potentials, when we compared both cervical MSC populations to MSCs from normal bone marrow (BM-MSCs, the gold standard). Interestingly, CeCa-MSCs were negative for the presence of human papilloma virus, indicating that these cells are not infected by such a viral agent. Also, interestingly, and in contrast to NCx-MSCs, CeCa-MSCs induced significant downregulation of surface HLA class I molecules (HLA-A*0201) on CaSki cells and other CeCa cell lines. We further observed that CeCa-MSCs inhibited antigen-specific T cell recognition of CaSki cells by cytotoxic T lymphocytes (CTLs). HLA class I downregulation on CeCa cells correlated with the production of IL-10 in cell cocultures. Importantly, this cytokine strongly suppressed recognition of CeCa cells by CTLs. In summary, this study demonstrates the presence of MSCs in CeCa and suggests that tumor-derived MSCs may provide immune protection to tumor cells by inducing downregulation of HLA class I molecules. This mechanism may have important implications in tumor growth.


Subject(s)
Cervix Uteri/cytology , Mesenchymal Stem Cells/metabolism , T-Lymphocytes, Cytotoxic/immunology , Uterine Cervical Neoplasms/metabolism , Antigens, Surface/metabolism , Bone Marrow Cells/metabolism , Cell Differentiation , Cell Proliferation , Cells, Cultured , Down-Regulation , Female , HLA-A2 Antigen/biosynthesis , HLA-A2 Antigen/metabolism , Humans , Interleukin-10/metabolism , Mesenchymal Stem Cells/cytology , Papillomaviridae , T-Lymphocytes, Cytotoxic/metabolism
14.
Exp Mol Pathol ; 89(2): 190-6, 2010 Oct.
Article in English | MEDLINE | ID: mdl-20599941

ABSTRACT

During carcinogenesis it is known that growth factors and cytokines from stromal and inflammatory cells from the microenvironment promote angiogenesis and lymphangiogenesis. However, the participation of macrophages and mast cells in these processes is not well understood. The aim of this study was to evaluate the relationship between mast cell and macrophage density with blood and lymphatic vessels in various stages of carcinoma of the uterine cervix. Tissue sections from archival paraffin-embedded samples from cases with cervical intraepithelial neoplasias (CIN) 1, 2, 3, carcinoma in situ, and invasive carcinoma were used. Immunohistochemical staining was done using the following antibodies: anti-LYVE-1; anti-CD31; anti-CD68, and anti-tryptase. Our results showed a significant increase in the number of macrophages in carcinoma in situ, a correlation between lymphatic vessels and macrophages in premalignant lesions CIN 2, and a correlation between mast cells and blood vessels in both CIN 2 and carcinoma in situ. In conclusion, our data underscore the importance of the recruitment of macrophages and mast cells in the development of tumor-associated blood and lymphatic capillaries.


Subject(s)
Carcinoma in Situ/immunology , Lymphangiogenesis/immunology , Macrophages/immunology , Mast Cells/immunology , Neovascularization, Pathologic/immunology , Uterine Cervical Dysplasia/immunology , Uterine Cervical Neoplasms/immunology , Uterine Cervical Neoplasms/pathology , Antigens, CD , Antigens, Differentiation, Myelomonocytic , Carcinoma in Situ/pathology , Case-Control Studies , Female , Humans , Macrophages/metabolism , Mast Cells/metabolism , Uterine Cervical Neoplasms/blood supply , Uterine Cervical Dysplasia/pathology
15.
Gac Med Mex ; 145(1): 51-60, 2009.
Article in Spanish | MEDLINE | ID: mdl-19256411

ABSTRACT

It is well-known that there are different tumor-type-dependent metastatic patterns. For example, in carcinomas metastatic spread is preferentially via the lymphatic system by which they reach regional lymph nodes through pre-existent afferent lymph vessels and/or newly formed lymph capillaries; while in sarcomas the favored pathway is through bloodvessels. These metastatic patterns have been used for many years by clinicians and surgeons for staging and tumor resection, particularly in the case of breast cancer. Recently this knowledge has been applied to detection and resection of sentinel lymph nodes. The lymphatic system drains the interstitial fluid from tissues and reincorporates it into the blood flow; in addition, it forms part of the host's immune defense and in pathological conditions, induces different types of lymph edema and participates in tumor invasion and metastasis. Although, the study of lymphangiogenesis was stagnated for several decades, it was not until a few years ago that biomolecular mechanisms were discovered and many specific markers are now in use to study the process of tumor dissemination and metastasis. There is a tendency to utilize molecular knowledge in clinical settings for grading and estimating prognostic significance of tumors as well as to develop specific therapeutic strategies.


Subject(s)
Lymphatic Metastasis , Neoplasms/pathology , Humans , Lymphatic System/anatomy & histology , Lymphatic System/physiology
16.
Gac. méd. Méx ; 145(1): 51-60, ene.-feb. 2009. ilus
Article in Spanish | LILACS | ID: lil-567733

ABSTRACT

Es bien sabido que existen diferentes patrones de metástasis dependiendo del tipo tumoral. Por ejemplo, la diseminación metastásica de los carcinomas es vía linfática preferencialmente, las células neoplásicas llegan a los ganglios linfáticos regionales a través de vasos linfáticos aferentes preexistentes o capilares linfáticos de nueva formación; en cambio, en los sarcomas la vía principal es a través de los vasos sanguíneos. Estos patrones metastásicos han sido utilizados durante muchos años por los clínicos y cirujanos para la etapificación y resección tumoral, particularmente en cáncer de mama. Recientemente este conocimiento ha sido aplicado para la detección y resección del ganglio centinela. El sistema linfático drena el líquido intersticial de los tejidos y lo reincorpora al sistema sanguíneo; además, forma parte de la defensa inmune del huésped y en condiciones patológicas induce diferentes tipos de linfedema y participa en la invasión y metástasis. El estudio de la linfangiogénesis permaneció aletargado por muchas décadas y no es sino hasta los últimos años que se han descrito mecanismos biomoleculares y marcadores específicos, los cuales actualmente se están utilizando para estudiar el proceso de diseminación tumoral y metástasis. Existe una tendencia hacia la aplicación clínica de este conocimiento molecular en la clínica para estimar el significado pronóstico de los tumores, así como para desarrollar estrategias terapéuticas específicas.


It is well-known that there are different tumor-type-dependent metastatic patterns. For example, in carcinomas metastatic spread is preferentially via the lymphatic system by which they reach regional lymph nodes through pre-existent afferent lymph vessels and/or newly formed lymph capillaries; while in sarcomas the favored pathway is through bloodvessels. These metastatic patterns have been used for many years by clinicians and surgeons for staging and tumor resection, particularly in the case of breast cancer. Recently this knowledge has been applied to detection and resection of sentinel lymph nodes. The lymphatic system drains the interstitial fluid from tissues and reincorporates it into the blood flow; in addition, it forms part of the host's immune defense and in pathological conditions, induces different types of lymph edema and participates in tumor invasion and metastasis. Although, the study of lymphangiogenesis was stagnated for several decades, it was not until a few years ago that biomolecular mechanisms were discovered and many specific markers are now in use to study the process of tumor dissemination and metastasis. There is a tendency to utilize molecular knowledge in clinical settings for grading and estimating prognostic significance of tumors as well as to develop specific therapeutic strategies.


Subject(s)
Humans , Lymphatic Metastasis , Neoplasms/pathology , Lymphatic System/anatomy & histology , Lymphatic System/physiology
SELECTION OF CITATIONS
SEARCH DETAIL
...