Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 94
Filtrar
1.
Exp Cell Res ; 442(2): 114188, 2024 Aug 14.
Artigo em Inglês | MEDLINE | ID: mdl-39128553

RESUMO

Cancer stem cells (CSC) are thought to be responsible for cancer phenotypes and cellular heterogeneity. Here we demonstrate that the human colon cancer cell line DLD1 contains two types of CSC-like cells that undergo distinct morphogenesis in the reconstituted basement membrane gel Matrigel. In our method with cancer cell spheroids, the parent cell line (DLD1-P) developed grape-like budding structures, whereas the other (DLD1-Wm) and its single-cell clones dynamically developed worm-like ones. Gene expression analysis suggested that the former mimicked intestinal crypt-villus morphogenesis, while the latter mimicked embryonic hindgut development. The organoids of DLD1-Wm cells rapidly extended in two opposite directions by expressing dipolar proteolytic activity. The invasive morphogenesis required the expression of MMP-2 and CD133 genes and ROCK activity. These cells also exhibited gastrula-like morphogenesis even in two-dimensional cultures without Matrigel. Moreover, the two DLD1 cell lines showed clear differences in cellular growth, tumor growth and susceptibility to paclitaxel. This study also provides a simple organoid culture method for human cancer cell lines. HT-29 and other cancer cell lines underwent characteristic morphogenesis in direct contact with normal fibroblasts. Such organoid cultures would be useful for investigating the nature of CSCs and for screening anti-cancer drugs. Our results lead to the hypothesis that CSC-like cells with both invasive activity and a fetal phenotype, i. e. oncofetal CSCs, are generated in some types of colon cancers.

2.
Cancer Sci ; 114(6): 2485-2498, 2023 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-36854451

RESUMO

Cancer tissues generally have molecular oxygen and serum component deficiencies because of poor vascularization. Recently, we revealed that ICAM1 is strongly activated through lipophagy in ovarian clear cell carcinoma (CCC) cells in response to starvation of long-chain fatty acids and oxygen and confers resistance to apoptosis caused by these harsh conditions. CD69 is a glycoprotein that is synthesized in immune cells and is associated with their activation through cellular signaling pathways. However, the expression and function of CD69 in nonhematological cells is unclear. Here, we report that CD69 is induced in CCC cells as in ICAM1. Mass spectrometry analysis of phosphorylated peptides followed by pathway analysis revealed that CD69 augments CCC cell binding to fibronectin (FN) in association with the phosphorylation of multiple cellular signaling molecules including the focal adhesion pathway. Furthermore, CD69 synthesized in CCC cells could facilitate cell survival because the CD69-FN axis can induce epithelial-mesenchymal transition. Experiments with surgically removed tumor samples revealed that CD69 is predominantly expressed in CCC tumor cells compared with other histological subtypes of epithelial ovarian cancer. Overall, our data suggest that cancer cell-derived CD69 can contribute to CCC progression through FN.


Assuntos
Fibronectinas , Neoplasias Ovarianas , Humanos , Feminino , Oxigênio , Neoplasias Ovarianas/patologia , Transdução de Sinais , Lipídeos , Linhagem Celular Tumoral
3.
Stem Cell Res Ther ; 13(1): 225, 2022 06 03.
Artigo em Inglês | MEDLINE | ID: mdl-35659728

RESUMO

BACKGROUND: Distinct subsets of cancer stem cells (CSCs) drive the initiation and progression of malignant tumors via enhanced self-renewal and development of treatment/apoptosis resistance. Endometrial CSC-selective drugs have not been successfully developed because most endometrial cell lines do not contain a sufficient proportion of stable CSCs. Here, we aimed to identify endometrial CSC-containing cell lines and to search for endometrial CSC-selective drugs. METHODS: We first assessed the presence of CSCs by identifying side populations (SPs) in several endometrial cancer cell lines. We then characterized cell viability, colony-formation, transwell invasion and xenotransplantion capability using the isolated SP cells. We also conducted real-time RT-PCR, immunoblot and immunofluorescence analyses of the cells' expression of CSC-associated markers. Focusing on 14 putative CSC-selective drugs, we characterized their effects on the proliferation and apoptosis of endometrial cancer cell lines, examining cell viability and annexin V staining. We further examined the inhibitory effects of the selected drugs, focusing on proliferation, invasion, expression of CSC-associated markers and tumor formation. RESULTS: We focused on HHUA cells, an endometrial cancer cell line derived from a well-differentiated endometrial adenocarcinoma. HHUA cells contained a sufficient proportion of stable CSCs with an SP phenotype (HHUA-SP). HHUA-SP showed greater proliferation, colony-formation, and invasive capabilities compared with the main population of HHUA cells (HHUA-MP). HHUA-SP generated larger tumors with higher expression of proliferation-related markers, Ki67, c-MYC and phosphorylated ERK compared with HHUA-MP when transplanted into immunodeficient mice. Among the 14 candidate drugs, sorafenib, an inhibitor of RAF pathways and multiple kinase receptors, inhibited cell proliferation and invasion in both HHUA-SP and -MP, but more profoundly in HHUA-SP. In vivo treatment with sorafenib for 4 weeks reduced the weights of HHUA-SP-derived tumors and decreased the expression of Ki67, ZEB1, and RAF1. CONCLUSIONS: Our results suggest that HHUA is a useful cell line for discovery and identification of endometrial CSC-selective drugs, and that sorafenib may be an effective anti-endometrial cancer drug targeting endometrial CSCs.


Assuntos
Neoplasias do Endométrio , Sistema de Sinalização das MAP Quinases , Animais , Carcinogênese/patologia , Linhagem Celular Tumoral , Proliferação de Células , Neoplasias do Endométrio/tratamento farmacológico , Neoplasias do Endométrio/genética , Neoplasias do Endométrio/metabolismo , Feminino , Humanos , Antígeno Ki-67/metabolismo , Camundongos , Células-Tronco Neoplásicas/metabolismo , Sorafenibe/metabolismo , Sorafenibe/farmacologia
4.
Cancer Sci ; 111(12): 4381-4392, 2020 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-32979884

RESUMO

Interaction of cancer cells with cancer-associated fibroblasts (CAFs) plays critical roles in tumor progression. Recently we proposed a new tumor invasion mechanism in which invasive cancer cells individually migrate on elongate protrusions of CAFs (CAF fibers) in 3-D collagen matrix. In this mechanism, cancer cells interact with fibronectin fibrils assembled on CAFs mainly through integrin-α5ß1. Here we tested whether this mechanism is applicable to the collective invasion of cancer cells, using two E-cadherin-expressing adenocarcinoma cell lines, DLD-1 (colon) and MCF-7 (breast). When hybrid spheroids of DLD-1 cells with CAFs were embedded into collagen gel, DLD-1 cells collectively but very slowly migrated through the collagen matrix in contact with CAFs. Epidermal growth factor and tumor necrosis factor-α promoted the collective invasion, possibly by reducing the E-cadherin junction, as did the transforming growth factor-ß inhibitor SB431542 by stimulating the outgrowth of CAFs. Transforming growth factor-ß itself inhibited the cancer cell invasion. Efficient collective invasion of DLD-1 cells required large CAF fibers or their assembly as stable adhesion substrates. Experiments with function-blocking Abs and siRNAs confirmed that DLD-1 cells adhered to fibronectin fibrils on CAFs mainly through integrin-α5ß1. Anti-E-cadherin Ab promoted the single cell invasion of DLD-1 cells by dissociating the E-cadherin junction. Although the binding affinity of MCF-7 cells to CAFs was lower than DLD-1, they also collectively invaded the collagen matrix in a similar fashion to DLD-1 cells. Our results suggest that the direct interaction with CAFs, as well as environmental cytokines, contributes to the collective invasion of cancers.


Assuntos
Fibroblastos Associados a Câncer/patologia , Colágeno/metabolismo , Fibroblastos/patologia , Fibronectinas/metabolismo , Integrina alfa5beta1/metabolismo , Invasividade Neoplásica/patologia , Células A549 , Adenocarcinoma/patologia , Amidas/farmacologia , Benzamidas/farmacologia , Fibroblastos Associados a Câncer/metabolismo , Adesão Celular , Linhagem Celular Tumoral , Movimento Celular , Cromonas/farmacologia , Neoplasias do Colo/metabolismo , Neoplasias do Colo/patologia , Tecido Conjuntivo/patologia , Dioxóis/farmacologia , Fator de Crescimento Epidérmico/metabolismo , Fibroblastos/metabolismo , Humanos , Imuno-Histoquímica/métodos , Células MCF-7 , Morfolinas/farmacologia , Invasividade Neoplásica/fisiopatologia , Piridinas/farmacologia , Esferoides Celulares/patologia , Fator de Crescimento Transformador beta/antagonistas & inibidores , Fator de Necrose Tumoral alfa/metabolismo
5.
F S Sci ; 1(1): 59-66, 2020 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-35559740

RESUMO

OBJECTIVE: To investigate the gene targets of estradiol (E2)-estrogen receptor-α (ESR1) in human endometrial stromal cells. DESIGN: Basic science. SETTING: University research center. PATIENT(S): Premenopausal women with or without endometriosis. INTERVENTION(S): Primary cultures of human endometrial stromal cells from healthy endometrium, with or without small-interfering RNA (siRNA) knockdown of ESR1 expression, were treated with E2 or vehicle control. MAIN OUTCOME MEASURE(S): Genome-wide RNA expression by RNA sequencing was compared in endometrial stromal cells with or without siRNA knockdown of ESR1 in the presence or absence of E2. Genome-wide recruitment of ESR1 to chromatin was assessed by chromatin immunoprecipitation sequencing. Gene expression by real-time qualitative polymerase chain reaction of a potential E2-ESR1 target gene was determined in endometrial stromal cells and endometriotic stromal cells. RESULT(S): We identified several important pathways that are dependent on E2-ESR1 signaling in endometrial stromal cells, including progesterone signaling, cell-matrix adhesion, and cytoskeleton rearrangement, as well as paracrine signaling by members of the fibroblast growth factor family. We detected a total of 709 ESR1 target sites on chromatin. By integrating data on genome-wide transcriptomic changes and E2-ESR1 binding sites, we identified inositol polyphosphate phosphatase type II (INPP4B) as a candidate E2-mediated suppressor of proliferation in healthy endometrial cells. INPP4B was downregulated in endometriosis-derived stromal cells. CONCLUSION(S): E2-ESR1 activates genes involved in human endometrial stromal cell cycle regulation, progesterone response, and production of stromal growth factors. Understanding the direct role of estrogen on the endometrial stroma and identifying downstream targets of E2-ESR1 can inform the development of targeted therapies for endometriosis and diminished endometrial receptivity.

6.
Endocr Rev ; 40(4): 1048-1079, 2019 08 01.
Artigo em Inglês | MEDLINE | ID: mdl-30994890

RESUMO

Pelvic endometriosis is a complex syndrome characterized by an estrogen-dependent chronic inflammatory process that affects primarily pelvic tissues, including the ovaries. It is caused when shed endometrial tissue travels retrograde into the lower abdominal cavity. Endometriosis is the most common cause of chronic pelvic pain in women and is associated with infertility. The underlying pathologic mechanisms in the intracavitary endometrium and extrauterine endometriotic tissue involve defectively programmed endometrial mesenchymal progenitor/stem cells. Although endometriotic stromal cells, which compose the bulk of endometriotic lesions, do not carry somatic mutations, they demonstrate specific epigenetic abnormalities that alter expression of key transcription factors. For example, GATA-binding factor-6 overexpression transforms an endometrial stromal cell to an endometriotic phenotype, and steroidogenic factor-1 overexpression causes excessive production of estrogen, which drives inflammation via pathologically high levels of estrogen receptor-ß. Progesterone receptor deficiency causes progesterone resistance. Populations of endometrial and endometriotic epithelial cells also harbor multiple cancer driver mutations, such as KRAS, which may be associated with the establishment of pelvic endometriosis or ovarian cancer. It is not known how interactions between epigenomically defective stromal cells and the mutated genes in epithelial cells contribute to the pathogenesis of endometriosis. Endometriosis-associated pelvic pain is managed by suppression of ovulatory menses and estrogen production, cyclooxygenase inhibitors, and surgical removal of pelvic lesions, and in vitro fertilization is frequently used to overcome infertility. Although novel targeted treatments are becoming available, as endometriosis pathophysiology is better understood, preventive approaches such as long-term ovulation suppression may play a critical role in the future.


Assuntos
Endometriose/etiologia , Animais , Endometriose/metabolismo , Endometriose/fisiopatologia , Endométrio/anormalidades , Feminino , Humanos , Camundongos , Receptores de Progesterona , Células Estromais/metabolismo , Células Estromais/fisiologia , Doenças Uterinas
7.
Sci Rep ; 9(1): 292, 2019 01 22.
Artigo em Inglês | MEDLINE | ID: mdl-30670761

RESUMO

Cancer-associated fibroblasts (CAFs) play critical roles in the tumor progression. However, it remains unclear how cancer cells migrate in the three-dimensional (3D) matrix of cancer tissues and how CAFs support the cancer invasion. Here we propose a novel mechanism of fibroblast-dependent cancer cell invasion in the 3D collagen matrix. Human cancer cell lines from the pancreas (Panc-1), lung (A549) and some other organs actively adhered to normal fibroblasts and primary lung CAFs in cultures. To show its significance in tumor invasion, we designed a new invasion assay in which homogeneous microspheroids consisting of cancer cells and fibroblasts were embedded into collagen gel. Time-lapse experiments showed that cancer cells adhered to and quickly migrated on the long protrusions of fibroblasts in the 3D collagen matrix. Fibroblast-free cancer cells poorly invaded the matrix. Experiments with function-blocking antibodies, siRNAs, and immunocytochemistry demonstrated that cancer cells adhered to fibroblasts through integrin α5ß1-mediated binding to fibronectin on the surface of fibroblasts. Immunochemical analyses of the co-cultures and lung cancers suggested that cancer cells could acquire the migratory force by the fibronectin/integrin signaling. Our results also revealed that the fibroblast-bound fibronectin was a preferential substrate for cancer cells to migrate in the collagen matrix.


Assuntos
Fibroblastos Associados a Câncer/patologia , Técnicas de Cultura de Células/métodos , Movimento Celular , Integrina alfa5beta1/metabolismo , Invasividade Neoplásica/patologia , Adesão Celular , Linhagem Celular Tumoral , Células Cultivadas , Técnicas de Cocultura , Colágeno , Humanos
8.
Biol Reprod ; 100(5): 1215-1227, 2019 05 01.
Artigo em Inglês | MEDLINE | ID: mdl-30649202

RESUMO

A decellularized uterine scaffold (DUS) prepared from rats permits recellularization and regeneration of uterine tissues when placed onto a partially excised uterus and supports pregnancy in a fashion comparable to the intact uterus. The underlying extracellular matrix (ECM) together with an acellular, perfusable vascular architecture preserved in DUS is thought to be responsible for appropriate regeneration of the uterus. To investigate this concept, we examined the effect of the orientation of the DUS-preserving ECM and the vascular architecture on uterine regeneration through placement of a DUS onto a partially defective uterine area in the reversed orientation such that the luminal face of the DUS was outside and the serosal face was inside. We characterized the tissue structure and function of the regenerated uterus, comparing the outcome to that when the DUS was placed in the correct orientation. Histological analysis revealed that aberrant structures including ectopic location of glands and an abnormal lining of smooth muscle layers were observed significantly more frequently in the reversed group than in the correct group (70% vs. 30%, P < 0.05). Despite the changes in tissue topology, the uteri regenerated with an incorrectly oriented DUS could achieve pregnancy in a way similar to uteri regenerated with a correctly oriented DUS. These results suggest that DUS-driven ECM orientation determines the regenerated uterus structure. Using DUS in the correct orientation is preferable when clinically applied. The disoriented DUS may deteriorate the tissue topology leading to structural disease of the uterus even though the fertility potential is not immediately affected.


Assuntos
Técnicas de Cultura de Células/métodos , Polaridade Celular/fisiologia , Matriz Extracelular/fisiologia , Regeneração/fisiologia , Alicerces Teciduais , Útero/citologia , Útero/fisiologia , Animais , Técnicas de Cultura de Células/veterinária , Células Cultivadas , Matriz Extracelular/química , Feminino , Intestino Delgado/citologia , Intestino Delgado/ultraestrutura , Gravidez , Ratos , Ratos Sprague-Dawley , Engenharia Tecidual/métodos , Engenharia Tecidual/veterinária , Alicerces Teciduais/química , Útero/ultraestrutura
9.
Stem Cell Reports ; 11(5): 1136-1155, 2018 11 13.
Artigo em Inglês | MEDLINE | ID: mdl-30392973

RESUMO

Defective endometrial stromal fibroblasts (EMSFs) contribute to uterine factor infertility, endometriosis, and endometrial cancer. Induced pluripotent stem cells (iPSCs) derived from skin or bone marrow biopsies provide a patient-specific source that can be differentiated to various cells types. Replacement of abnormal EMSFs is a potential novel therapeutic approach for endometrial disease; however, the methodology or mechanism for differentiating iPSCs to EMSFs is unknown. The uterus differentiates from the intermediate mesoderm (IM) to form coelomic epithelium (CE) followed by the Müllerian duct (MD). Here, we successfully directed the differentiation of human iPSCs (hiPSCs) through IM, CE, and MD to EMSFs under molecularly defined embryoid body culture conditions using specific hormonal treatments. Activation of CTNNB1 was essential for expression of progesterone receptor that mediated the final differentiation step of EMSFs before implantation. These hiPSC-derived tissues illustrate the potential for iPSC-based endometrial regeneration for future cell-based treatments.


Assuntos
Endométrio/citologia , Fibroblastos/citologia , Células-Tronco Pluripotentes Induzidas/citologia , Progesterona/farmacologia , Via de Sinalização Wnt , beta Catenina/metabolismo , Diferenciação Celular/efeitos dos fármacos , Diferenciação Celular/genética , Células Cultivadas , Decídua/metabolismo , Células Epiteliais/citologia , Células Epiteliais/efeitos dos fármacos , Células Epiteliais/metabolismo , Feminino , Fibroblastos/efeitos dos fármacos , Fibroblastos/metabolismo , Humanos , Células-Tronco Pluripotentes Induzidas/efeitos dos fármacos , Células-Tronco Pluripotentes Induzidas/metabolismo , Mesoderma/citologia , Ductos Paramesonéfricos/citologia , Linha Primitiva/citologia , Células Estromais/citologia , Células Estromais/efeitos dos fármacos , Células Estromais/metabolismo , Transcriptoma/genética , Via de Sinalização Wnt/efeitos dos fármacos
10.
Oncotarget ; 8(45): 78312-78326, 2017 Oct 03.
Artigo em Inglês | MEDLINE | ID: mdl-29108231

RESUMO

Extracellular acidity is a hallmark of solid tumors and is associated with metastasis in the tumor microenvironment. Acidic extracellular pH (pH e ) has been found to increase intracellular Ca2+ and matrix metalloproteinase-9 (MMP-9) expression by activating NF-κB in the mouse B16 melanoma model. The present study assessed whether TRPM5, an intracellular Ca2+-dependent monovalent cation channel, is associated with acidic pH e signaling and induction of MMP-9 expression in this mouse melanoma model. Treatment of B16 cells with Trpm5 siRNA reduced acidic pH e -induced MMP-9 expression. Enforced expression of Trpm5 increased the rate of acidic pH e -induced MMP-9 expression, as well as increasing experimental lung metastasis. This genetic manipulation did not alter the pH e critical for MMP-9 induction but simply amplified the percentage of inducible MMP-9 at each pH e . Treatment of tumor bearing mice with triphenylphosphine oxide (TPPO), an inhibitor of TRPM5, significantly reduced spontaneous lung metastasis. In silico analysis of clinical samples showed that high TRPM5 mRNA expression correlated with poor overall survival rate in patients with melanoma and gastric cancer but not in patients with cancers of the ovary, lung, breast, and rectum. These results showed that TRPM5 amplifies acidic pH e signaling and may be a promising target for preventing metastasis of some types of tumor.

11.
Cancer Sci ; 107(12): 1909-1918, 2016 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-27685891

RESUMO

The laminin γ2 chain, a subunit of laminin-332 (α3ß3γ2), is a molecular marker for invasive cancer cells, but its pathological roles in tumor progression remain to be clarified. It was recently found that the most N-terminal, domain V (dV) of γ2 chain has activities to bind CD44 and stimulate tumor cell migration and vascular permeability. In the present study, we prepared a mAb recognizing γ2 dV. Immunoblotting with this antibody, for the first time, showed that proteolytic fragments containing dV in a range of 15-80 kDa were highly produced in various human cancer cell lines and lung cancer tissues. In immunohistochemistry of adenocarcinomas and squamous cell carcinomas of the lung, this antibody immunostained the cytoplasm of invasive tumor cells and adjacent stroma much more strongly than a widely used antibody recognizing the C-terminal core part of the processed γ2 chain. This suggests that the dV fragments are highly accumulated in tumor cells and stroma compared to the processed γ2 protein. The strong tumor cell staining with the dV antibody correlated with the tumor malignancy grade. We also found that the laminin ß3 and α3 chains were frequently overexpressed in tumor cells and tumor stroma, respectively. The cytoplasmic dV detection was especially prominent in tumor cells infiltrating stroma, but low in the cells surrounded by basement membranes, suggesting that the active tumor-stroma interaction is critical for the aberrant γ2 expression. The present study suggests important roles of laminin γ2 N-terminal fragments in tumor progression.


Assuntos
Laminina/metabolismo , Neoplasias Pulmonares/metabolismo , Animais , Anticorpos Monoclonais , Biomarcadores , Linhagem Celular Tumoral , Humanos , Imuno-Histoquímica , Laminina/química , Neoplasias Pulmonares/patologia , Camundongos , Domínios e Motivos de Interação entre Proteínas , Transporte Proteico
12.
Fertil Steril ; 106(2): 487-496.e1, 2016 08.
Artigo em Inglês | MEDLINE | ID: mdl-27068301

RESUMO

OBJECTIVE: To create a bioengineered uterine patch for uterine repair of a partially defect uterus. DESIGN: Three different decellularized uterine scaffolds were recellularized in vitro with primary uterine cells and green fluorescent protein- (GPF-) labeled bone marrow-derived mesenchymal stem cells (GFP-MSCs). The patches were transplanted in vivo to investigate their tissue adaptation and supporting capacity during pregnancy. SETTING: Research laboratory. ANIMAL(S): Female Lewis rats (n = 9) as donors to generate whole-uterus scaffolds using three different protocols (n = 3 per protocol); Sprague Dawley rats (n = 40) for primary uterus cell isolation procedures (n = 10) and for transplantation/pregnancy studies (n = 30); and male Sprague Dawley rats (n = 12) for mating. INTERVENTION(S): Decellularization was achieved by whole-uterus perfusion with buffered or nonbuffered Triton-X100 and dimethyl sulfoxide (DMSO; group P1/P2) or with sodium deoxycholate (group P3). Primary uterine cells and GFP-MSCs were used to develop uterine tissue constructs, which were grafted to uteri with partial tissue defects. MAIN OUTCOME MEASURE(S): Recellularization efficiency and graft quality were analyzed morphologically, immunohistochemically, and by real-time quantitative polymerase chain reaction (PCR). The location and number of fetuses were documented during pregnancy days 16-20. RESULT(S): Pregnancy and fetal development were normal in groups P1 and P2, with fetal development over patched areas. Group P3 showed significant reduction of fetal numbers, and embryos were not seen in the grafted area. Quantitative PCR and immunohistochemistry revealed uterus-like tissue in the patches, which had been further reconstructed by infiltrating host cells after transplantation. CONCLUSION(S): Primary uterine cells and MSCs can be used to reconstruct decellularized uterine tissue. The bioengineered patches made from triton-X100+DMSO-generated scaffolds were supportive during pregnancy. These protocols should be explored further to develop suitable grafting material to repair partially defect uteri and possibly to create a complete bioengineered uterus.


Assuntos
Transplante de Células-Tronco Mesenquimais , Células-Tronco Mesenquimais/fisiologia , Engenharia Tecidual/métodos , Alicerces Teciduais , Útero/transplante , Animais , Células Cultivadas , Feminino , Desenvolvimento Fetal , Regulação da Expressão Gênica no Desenvolvimento , Genes Reporter , Idade Gestacional , Proteínas de Fluorescência Verde/biossíntese , Proteínas de Fluorescência Verde/genética , Tamanho da Ninhada de Vivíparos , Células-Tronco Mesenquimais/metabolismo , Gravidez , Cultura Primária de Células , RNA Mensageiro/genética , RNA Mensageiro/metabolismo , Ratos Endogâmicos Lew , Transfecção , Útero/metabolismo
13.
Semin Reprod Med ; 33(5): 366-72, 2015 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-26285168

RESUMO

Regenerative medicine offers the potential for replacement or repair of different types of cells within damaged tissues or the tissues themselves, typically through cell therapy or tissue engineering. Stem cells are critical to these approaches; indeed, the involvement of bone marrow in the differentiation of stem cells to nonhematopoietic cells is well demonstrated. Further, the contribution of bone marrow-derived stem cells in promoting neoangiogenesis has been demonstrated not only in animal models, but also in human clinical trials with an excellent safety profile. Recent evidence indicates that the endometrium is a tissue with the potential for regeneration through such approaches. The presence of donor cells in the endometrium of women receiving bone marrow transplantation suggests a hematopoietic source with the ability to renew this tissue. Here we describe the role of cell therapy with bone marrow-derived stem cells in treating endometrial dysfunction in Asherman syndrome and/or endometrial atrophy in human and murine models. Additionally, the emerging field of tissue engineering has recently been applied in the reproductive tissues-beyond the endometrium-with elegant studies involving humans and animal models.


Assuntos
Transplante de Medula Óssea , Ginatresia/terapia , Transplante de Células-Tronco , Engenharia Tecidual , Doenças Uterinas/terapia , Útero , Vagina , Antígeno AC133 , Animais , Antígenos CD , Atrofia , Feminino , Glicoproteínas , Humanos , Camundongos , Peptídeos , Procedimentos de Cirurgia Plástica , Medicina Regenerativa , Doenças Uterinas/patologia
14.
Biol Reprod ; 93(4): 84, 2015 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-26316062

RESUMO

Uterine endometrium is one of the most important organs for species preservation. However, the physiology of human endometrium remains poorly understood, because the human endometrium undergoes rapid and large changes during each menstrual cycle and it is very difficult to investigate human endometrium as one organ. This remarkable regenerative capacity of human endometrium strongly suggests the existence of adult stem cells, and physiology of endometrium cannot be explained without adult stem cells. Therefore, investigating endometrial stem/progenitor cells should lead to a breakthrough in understanding the normal endometrial physiology and the pathophysiology of endometrial neoplastic disorders, such as endometriosis and endometrial cancer. Several cell populations have been discovered as putative endometrial stem/progenitor cells. Emerging evidence reveals that the endometrial side population (SP) is one of the potential endometrial stem/progenitor populations. Of all the endometrial stem/progenitor cell candidates, the endometrial SP (ESP) is best investigated in vitro and in vivo, and has the largest number of references. In this review, we provide an overview of the accumulating evidence for the ESP cells, both directly from human endometria and from cultured endometrial cells. Furthermore, SP cells are compared to other potential stem/progenitor cells, and we discuss their stem cell properties. We also discuss the difficulties and unsolved issues in endometrial stem cell biology.


Assuntos
Endométrio/citologia , Células da Side Population/fisiologia , Células-Tronco/fisiologia , Animais , Feminino , Humanos
15.
Biol Reprod ; 93(2): 37, 2015 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-26108791

RESUMO

Repeated and dramatic pregnancy-induced uterine enlargement and remodeling throughout reproductive life suggests the existence of uterine smooth muscle stem/progenitor cells. The aim of this study was to isolate and characterize stem/progenitor-like cells from human myometrium through identification of specific surface markers. We here identify CD49f and CD34 as markers to permit selection of the stem/progenitor cell-like population from human myometrium and show that human CD45(-) CD31(-) glycophorin A(-) and CD49f(+) CD34(+) myometrial cells exhibit stem cell-like properties. These include side population phenotypes, an undifferentiated status, high colony-forming ability, multilineage differentiation into smooth muscle cells, osteoblasts, adipocytes, and chondrocytes, and in vivo myometrial tissue reconstitution following xenotransplantation. Furthermore, CD45(-) CD31(-) glycophorin A(-) and CD49f(+) CD34(+) myometrial cells proliferate under hypoxic conditions in vitro and, compared with the untreated nonpregnant myometrium, show greater expansion in the estrogen-treated nonpregnant myometrium and further in the pregnant myometrium in mice upon xenotransplantation. These results suggest that the newly identified myometrial stem/progenitor-like cells influenced by hypoxia and sex steroids may participate in pregnancy-induced uterine enlargement and remodeling, providing novel insights into human myometrial physiology.


Assuntos
Antígenos CD34/genética , Antígenos CD34/fisiologia , Integrina alfa6/genética , Integrina alfa6/fisiologia , Miométrio/metabolismo , Células-Tronco/fisiologia , Útero/fisiologia , Animais , Diferenciação Celular , Hipóxia Celular , Linhagem da Célula/genética , Feminino , Glicoforinas/biossíntese , Glicoforinas/genética , Células-Tronco Hematopoéticas , Humanos , Camundongos , Miométrio/citologia , Molécula-1 de Adesão Celular Endotelial a Plaquetas/biossíntese , Molécula-1 de Adesão Celular Endotelial a Plaquetas/genética , Gravidez
16.
Clin Exp Metastasis ; 32(5): 405-15, 2015 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-25990436

RESUMO

Laminin γ2 (Lmγ2) chain, a subunit of the basement membrane protein laminin-332, is regarded as a typical cancer invasion marker. The overexpression of Lmγ2 chain by invasive cancer cells correlates with poor prognosis of cancer patients, and its forced expression in human cancer cells promotes their invasive growth in a nude mouse model. However, its actual roles in cancer progression, as well as the mechanism of its proinvasive effect, remain unclear. CD44 is known to be an important cancer stem cell marker and support cancer progression and stem cell functions. Here we demonstrate that amino-terminal fragments of Lmγ2 interact with CD44 on the membrane of breast cancer cells. Lmγ2 highly bound to the metastatic cell line MDA-MB-231 but poorly to the benign cell line MCF-7. The membrane receptor for Lmγ2 on MDA-MB-231 cells was identified to be the standard form of CD44 (CD44s) by co-immunoprecipitation, affinity chromatography and direct protein interaction assay. Lmγ2 interacted with CD44s through EGF-like repeat 2/3 in the Lmγ2 amino-terminus. Amino-terminal fragments of Lmγ2 induced the phosphorylation of CD44 cytoplasmic domain and stimulated migration of the cancer cells in a CD44-dependent manner. This migration was blocked by inhibitors of TGF-ß receptor I (TGF-ßRI) kinase. These results suggest that two important tumor markers, Lmγ2 and CD44, cooperate for cancer progression and possibly for cancer stem cell functions. TGF-ßRI may be involved in the Lmγ2/CD44 interaction.


Assuntos
Neoplasias da Mama/metabolismo , Neoplasias da Mama/secundário , Movimento Celular , Receptores de Hialuronatos/metabolismo , Laminina/metabolismo , Células-Tronco Neoplásicas/metabolismo , Proteínas Serina-Treonina Quinases/metabolismo , Receptores de Fatores de Crescimento Transformadores beta/metabolismo , Animais , Western Blotting , Adesão Celular , Proliferação de Células , Cromatografia de Afinidade , Feminino , Humanos , Imunoprecipitação , Camundongos , Invasividade Neoplásica , Células-Tronco Neoplásicas/patologia , Fosforilação , Receptor do Fator de Crescimento Transformador beta Tipo I , Transdução de Sinais , Células Tumorais Cultivadas
17.
Endocr J ; 62(1): 93-9, 2015.
Artigo em Inglês | MEDLINE | ID: mdl-25312800

RESUMO

The objective of this study was to assess the potential predictive factors for follicle growth, ovulation, and pregnancy rate in patients with primary ovarian insufficiency/premature ovarian failure (POI/POF). We enrolled 25 POI patients with desired fertility who were treated and monitored for a minimum of 7 months between the years of 2000-2009 into this retrospective study. The clinical, endocrinologic, chromosomal, and autoimmunologic parameters of these patients were collected. Furthermore, hormonal backgrounds on each of 620 treatment cycles were investigated. The main outcome measures were follicle growth, ovulation, and pregnancy rate. Four of 25 patients (16%) conceived while being monitored and undergoing treatment. Follicle growth, ovulation, and pregnancy rate were not significantly different as a function of parity, iatrogenic history (e.g., chemotherapy), age of disease onset, serum estradiol (E(2))/follicle stimulating hormone (FSH) level at the time of diagnosis, chromosomal abnormality, and positive autoantibody titer. The serum E2 levels on days 1-5 of withdrawal bleeding (Day 1-5 E(2)) were significantly higher in the cycles with successful follicle growth and ovulation than unsuccessful cycles (P<0.05). Receiver-operator characteristic curve analysis revealed the cut-off value of the Day 1-5 E(2) to be 15.5 pg/mL, and an area under the curve (AUC) value of 0.674 for follicle growth and 0.752 for ovulation. The results suggest that cycles with a Day 1-5 E(2)≥15.5 pg/mL have a higher rate of follicle growth and ovulation in patients with POI.


Assuntos
Estradiol/sangue , Infertilidade Feminina/etiologia , Metrorragia/etiologia , Ovário/fisiopatologia , Insuficiência Ovariana Primária/sangue , Adulto , Biotransformação , Estrogênios/farmacocinética , Estrogênios/uso terapêutico , Feminino , Fármacos para a Fertilidade Feminina/farmacocinética , Fármacos para a Fertilidade Feminina/uso terapêutico , Seguimentos , Humanos , Infertilidade Feminina/prevenção & controle , Japão/epidemiologia , Metrorragia/prevenção & controle , Oogênese/efeitos dos fármacos , Ovário/efeitos dos fármacos , Ovário/metabolismo , Ovulação/efeitos dos fármacos , Gravidez , Taxa de Gravidez , Insuficiência Ovariana Primária/tratamento farmacológico , Insuficiência Ovariana Primária/fisiopatologia , Curva ROC , Estudos Retrospectivos , Adulto Jovem
18.
Biomaterials ; 35(31): 8791-8800, 2014 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-25043501

RESUMO

Despite dramatic progress in infertility treatments and assisted reproduction, no effective therapies exist for complete loss of uterine structure and/or function. For such patients, genetic motherhood is possible only through gestational surrogacy or uterine transplantation. However, many ethical, social, technical and safety challenges accompany such approaches. A theoretical alternative is to generate a bioartificial uterus, which requires engineering of uterine architecture and appropriate cellular constituents. Here, rat uteri decellularization by aortic perfusion with detergents produced an underlying extracellular matrix together with an acellular, perfusable vascular architecture. Uterine-like tissues were then regenerated and maintained in vitro for up to 10 d through decellularized uterine matrix (DUM) reseeding with adult and neonatal rat uterine cells and rat mesenchymal stem cells followed by aortic perfusion in a bioreactor. Furthermore, DUM placement onto a partially excised uterus yielded recellularization and regeneration of uterine tissues and achievement of pregnancy nearly comparable to the intact uterus. These results suggest that DUM could be used for uterine regeneration, and provides insights into treatments for uterine factor infertility.


Assuntos
Matriz Extracelular/transplante , Regeneração , Alicerces Teciduais/química , Útero/citologia , Útero/fisiologia , Animais , Células Cultivadas , Detergentes/química , Matriz Extracelular/química , Matriz Extracelular/ultraestrutura , Feminino , Técnicas de Cultura de Órgãos/métodos , Gravidez , Ratos , Ratos Endogâmicos F344 , Engenharia Tecidual/métodos , Útero/transplante , Útero/ultraestrutura
19.
FEBS J ; 281(15): 3346-56, 2014 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-24903600

RESUMO

Matrix metalloproteinase (MMP)-7 binds to cell surface cholesterol sulfate (CS) and acts as a membrane-associated protease. We have previously found that CS modulates the substrate preference of MMP-7, thereby regulating its pericellular proteolytic action. MMP-7 potentially associates with the cell surface via sulfatide (SM4) and cardiolipin (CL) when they are overexpressed on the cell surface. Here, we investigated the molecular interaction between these acidic lipids and MMP-7 or its substrates, and their effects on the activity of MMP-7. Studies using MMP-7 variants with low CS-binding ability suggested that these lipids interact with a similar site on MMP-7. The hydroxamate-based MMP inhibitor TAPI-1 markedly reduced the affinity of MMP-7 for CS and CL, whereas that for SM4 was not affected by TAPI-1. These three acidic lipids also had different effects on the hydrolytic activity of MMP-7 towards a small peptide substrate: SM4, CL and CS reduced the activity to 80%, 92%, and 20%, respectively. Nevertheless, SM4 and CS similarly accelerated the MMP-7-catalyzed degradation of fibronectin and laminin-332, whereas CL did not. The increased proteolysis of substrate was observed only when both substrate and enzyme had affinity for the lipid, suggesting that the lipids probably bring the reactants into closer proximity. Furthermore, MMP-7 bound to cell surface SM4 or CS cleaved specific cell surface proteins and released similar fragments, whereas the cleavage was not stimulated by cell surface CL-bound MMP-7. This study provides a novel mechanism by which acidic lipids differentially regulate pericellular proteolysis by MMP-7 through allosteric alteration of the substrate-binding site and their inherent affinities for MMP-7 substrates.


Assuntos
Cardiolipinas/metabolismo , Ésteres do Colesterol/metabolismo , Metaloproteinase 7 da Matriz/metabolismo , Sulfoglicoesfingolipídeos/metabolismo , Ligação Competitiva , Cardiolipinas/química , Moléculas de Adesão Celular/química , Linhagem Celular Tumoral , Membrana Celular/enzimologia , Ésteres do Colesterol/química , Dipeptídeos/química , Dipeptídeos/farmacologia , Fibronectinas/química , Humanos , Ácidos Hidroxâmicos/química , Ácidos Hidroxâmicos/farmacologia , Cinética , Metaloproteinase 7 da Matriz/química , Inibidores de Metaloproteinases de Matriz/química , Inibidores de Metaloproteinases de Matriz/farmacologia , Ligação Proteica , Proteólise , Sulfoglicoesfingolipídeos/química , Calinina
20.
Exp Cell Res ; 326(2): 267-79, 2014 Aug 15.
Artigo em Inglês | MEDLINE | ID: mdl-24780821

RESUMO

Interaction between tumor cells and stromal fibroblasts plays essential roles in tumor progression. However, its detailed molecular mechanism remains unclear. To understand the mechanism, we investigated molecules mediating this interaction using the three-dimensional (3D) co-culture system of Panc-1 pancreatic carcinoma cells with normal fibroblasts. When the two kinds of cells were placed on the top of collagen gel, the tumor cells scattered into the fibroblast layer, apparently undergoing epithelial-mesenchymal transition. When fibroblasts were placed within collagen gel, Panc-1 cells actively invaded into the collagen gel, extending a microtubule-based long protrusion. Although transforming growth factor-ß (TGF-ß) and hepatocyte growth factor (HGF) individually stimulated the tumor cell invasion into collagen gel without fibroblasts, TGF-ß signaling inhibitors (SB431542 and LY2157299) significantly enhanced the Panc-1 cell invasion in the 3D co-culture with fibroblasts. Experiments with HGF/Met signaling inhibitors or with the fibroblast conditioned medium revealed that HGF was a major invasion-promoting factor secreted from fibroblasts and SB431542 increased the HGF secretion by blocking the HGF-suppressing activity of cancer cell-derived TGF-ß. These results indicate that HGF and TGF-ß are critical regulators for both tumor-stroma interaction and tumor invasion. The results also suggest that TGF-ß signaling inhibitors may promote tumor progression under some pathological conditions.


Assuntos
Fator de Crescimento de Hepatócito/metabolismo , Invasividade Neoplásica/fisiopatologia , Neoplasias Pancreáticas/metabolismo , Fator de Crescimento Transformador beta/antagonistas & inibidores , Benzamidas/farmacologia , Linhagem Celular , Linhagem Celular Tumoral , Técnicas de Cocultura , Colágeno/metabolismo , Dioxóis/farmacologia , Transição Epitelial-Mesenquimal , Fibroblastos/metabolismo , Fator de Crescimento de Hepatócito/antagonistas & inibidores , Humanos , Modelos Estatísticos , Invasividade Neoplásica/patologia , Neoplasias Pancreáticas/patologia , Transdução de Sinais , Células Estromais/metabolismo , Fator de Crescimento Transformador beta/metabolismo
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA