Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 43
Filtrar
1.
Cell Stress Chaperones ; 28(6): 835-846, 2023 11.
Artículo en Inglés | MEDLINE | ID: mdl-37632625

RESUMEN

Small heat shock proteins (sHSPs) play a crucial role under abiotic stress and are present in all organisms, from eukaryotes to prokaryotes. However, studies on the sHSP gene family in red alga are limited. In this study, we aimed to identify and characterize NysHSP genes from the genome of N. yezoensis, a marine red alga adapted to the stressful intertidal zone. We identified seven NysHSP genes distributed on all three chromosomes. Expression analysis revealed that all NysHSP genes responded to H2O2 and heat stress in the gametophytic thalli, but these genes responded only to heat stress in the sporophytic conchocelis. NysHSP20.3, which has an acidic isoelectric point (pI) and short N-terminal region, was localized as granules in the cytosol. Fluorescence imaging of the NysHSP25.8-GFP and NysHSP28.4-GFP fusion proteins revealed that these proteins were located in the chloroplast. Based on their characteristics and cellular localization, the NysHSPs are divided into two subfamilies. Subfamily I includes four sHSP genes that strongly respond to heat stress and encode a protein localized in the cytosol. The NysHSP gene of subfamily II encodes a polypeptide with a long N-terminal region located in the chloroplast. This study provides insights into the evolution and function of the sHSP gene family of the marine red alga N. yezoensis and how it adapts to the stressful intertidal zone.


Asunto(s)
Proteínas de Choque Térmico Pequeñas , Rhodophyta , Proteínas de Choque Térmico Pequeñas/genética , Proteínas de Choque Térmico Pequeñas/metabolismo , Peróxido de Hidrógeno/metabolismo , Cloroplastos/genética , Cloroplastos/metabolismo , Rhodophyta/genética
2.
Mol Biol Rep ; 50(10): 8459-8467, 2023 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-37632632

RESUMEN

BACKGROUND: Intestinal injury is one of the main side-effects of cisplatin chemotherapy, impairing the quality of life in patients with cancer. In this study, we investigated the protective effects of recombinant soluble thrombomodulin (rsTM), which is a potent anti-inflammatory agent, on cisplatin-induced intestinal injury. METHODS: We first evaluated the effects of rsTM on intestinal injury caused by cisplatin in mice in vivo. Disease progression was monitored by analyzing loss of body weight and histological changes in intestinal tissue. We then investigated the effects of rsTM on mouse intestinal organoid formation and growth in vitro. Gene expression levels were analyzed by quantitative real-time polymerase chain reaction and Western blotting. RESULTS: rsTM treatment significantly attenuated the loss of body weight, histological damage and gene expression levels of pro-inflammatory cytokines such as interleukin-6, tumor necrosis factor-α and high-mobility group box-1 in a cisplatin-treated mouse model. Furthermore, rsTM alleviated the inflammatory response and apoptosis in a cisplatin-treated intestinal epithelial organoid model. CONCLUSION: rsTM suppresses cisplatin-induced intestinal epithelial cell-derived cytokine production and alleviates intestinal mucositis.


Asunto(s)
Cisplatino , Citocinas , Humanos , Ratones , Animales , Citocinas/metabolismo , Cisplatino/efectos adversos , Trombomodulina/genética , Calidad de Vida , Células Epiteliales/metabolismo , Peso Corporal
3.
Cells ; 12(5)2023 02 24.
Artículo en Inglés | MEDLINE | ID: mdl-36899862

RESUMEN

Sepsis is a systemic inflammatory disorder that leads to the dysfunction of multiple organs. In the intestine, the deregulation of the epithelial barrier contributes to the development of sepsis by triggering continuous exposure to harmful factors. However, sepsis-induced epigenetic changes in gene-regulation networks within intestinal epithelial cells (IECs) remain unexplored. In this study, we analyzed the expression profile of microRNAs (miRNAs) in IECs isolated from a mouse model of sepsis generated via cecal slurry injection. Among 239 miRNAs, 14 miRNAs were upregulated, and 9 miRNAs were downregulated in the IECs by sepsis. Upregulated miRNAs in IECs from septic mice, particularly miR-149-5p, miR-466q, miR-495, and miR-511-3p, were seen to exhibit complex and global effects on gene regulation networks. Interestingly, miR-511-3p has emerged as a diagnostic marker in this sepsis model due to its increase in blood in addition to IECs. As expected, mRNAs in the IECs were remarkably altered by sepsis; specifically, 2248 mRNAs were decreased, while 612 mRNAs were increased. This quantitative bias may be possibly derived, at least partly, from the direct effects of the sepsis-increased miRNAs on the comprehensive expression of mRNAs. Thus, current in silico data indicate that there are dynamic regulatory responses of miRNAs to sepsis in IECs. In addition, the miRNAs that were increased with sepsis had enriched downstream pathways including Wnt signaling, which is associated with wound healing, and FGF/FGFR signaling, which has been linked to chronic inflammation and fibrosis. These modifications in miRNA networks in IECs may lead to both pro- and anti-inflammatory effects in sepsis. The four miRNAs discovered above were shown to putatively target LOX, PTCH1, COL22A1, FOXO1, or HMGA2, via in silico analysis, which were associated with Wnt or inflammatory pathways and selected for further study. The expressions of these target genes were downregulated in sepsis IECs, possibly through posttranscriptional modifications of these miRNAs. Taken together, our study suggests that IECs display a distinctive miRNA profile which is capable of comprehensively and functionally reshaping the IEC-specific mRNA landscape in a sepsis model.


Asunto(s)
MicroARNs , Sepsis , Ratones , Animales , Perfilación de la Expresión Génica , MicroARNs/genética , Células Epiteliales/metabolismo , Intestinos , Sepsis/genética
4.
J Microbiol Biotechnol ; 33(3): 310-318, 2023 Mar 28.
Artículo en Inglés | MEDLINE | ID: mdl-36734134

RESUMEN

Microalgae are attracting much attention as promising, eco-friendly producers of bioenergy due to their fast growth, absorption of carbon dioxide from the atmosphere, and production capacity in wastewater and salt water. However, microalgae can only accumulate large quantities of lipid in abiotic stress, which reduces productivity by decreasing cell growth. In this study, the strategy was investigated to increase cell viability and lipid production by overexpressing S-adenosylmethionine (SAM) synthetase (SAMS) in the microalga Chlamydomonas reinhardtii. SAM is a substance that plays an important role in various intracellular biochemical reactions, such as cell proliferation and stress response, and the overexpression of SAMS could allow cells to withstand the abiotic stress and increase productivity. Compared to wild-type C. reinhardtii, recombinant cells overexpressing SAMS grew 1.56-fold faster and produced 1.51-fold more lipids in a nitrogen-depleted medium. Furthermore, under saline-stress conditions, the survival rate and lipid accumulation were 1.56 and 2.04 times higher in the SAMS-overexpressing strain, respectively. These results suggest that the overexpression of SAMS in recombinant C. reinhardtii has high potential in the industrial-scale production of biofuels and various other high-value-added materials.


Asunto(s)
Chlamydomonas reinhardtii , Chlamydomonas , Lípidos , Metionina Adenosiltransferasa , Chlamydomonas reinhardtii/química , Proliferación Celular
5.
Cancers (Basel) ; 14(13)2022 Jul 04.
Artículo en Inglés | MEDLINE | ID: mdl-35805039

RESUMEN

Breast cancer is the most common cancer in women worldwide, and lung metastasis is one of the most frequent distant metastases. When breast cancer metastasizes to the lung, group 2 innate lymphoid cells (ILC2s) are thought to promote tumor growth via the activation of myeloid-derived suppressor cells (MDSCs), which are known to negatively regulate anticancer immune responses. However, it remains to be elucidated exactly how this ILC2-MDSC interaction is involved in tumor growth during metastases formation. Using a 4T1/LM4 breast cancer mouse model, we found that ILC2s were activated in both the micro- and macrometastatic regions, suggesting sustained activation throughout the metastatic cascades via IL-33/ST2 signaling. Consistent with IL-13 secretion from activated ILC2s, the frequencies of polymorphonuclear (PMN)- and monocytic (M)-MDSCs were also significantly elevated during the progression from micro- to macrometastatic cancer. However, the effects of ILC2-induced MDSC functionality on the microenvironment differed in a metastatic-stage-specific manner. Our findings indicate that ILC2s may induce the immunosuppressive functions of MDSCs during the later stages of metastasis. Concomitantly, ILC2 may instigate extracellular matrix remodeling by PMN-MDSC activation during the early stages of metastasis. These metastatic-stage-specific changes may contribute to metastatic tumor growth in the microenvironment of breast cancer lung metastasis.

6.
Sci Rep ; 11(1): 21597, 2021 11 03.
Artículo en Inglés | MEDLINE | ID: mdl-34732818

RESUMEN

The ability of integrins on the cell surface to mediate cell adhesion to the extracellular matrix ligands is regulated by intracellular signaling cascades. During this signaling process, the talin (TLN) recruited to integrin cytoplasmic tails plays the critical role of the major adaptor protein to trigger integrin activation. Thus, intracellular levels of TLN are thought to determine integrin-mediated cellular functions. However, the epigenetic regulation of TLN expression and consequent modulation of integrin activation remain to be elucidated. Bioinformatics analysis led us to consider miR-200c-3p as a TLN1-targeting miRNA. To test this, we have generated miR-200c-3p-overexpressing and miR-200c-3p-underexpressing  cell lines, including HEK293T, HCT116, and LNCaP cells. Overexpression of miR-200c-3p resulted in a remarkable decrease in the expression of TLN1, which was associated with the suppression of integrin-mediated cell adhesion to fibronectin. In contrast, the reduction in endogenous miR-200c-3p levels led to increased expression of TLN1 and enhanced cell adhesion to fibronectin and focal adhesion plaques formation. Moreover, miR-200c-3p was found to target TLN1 by binding to its 3'-untranslated region (UTR). Taken together, our data indicate that miR-200c-3p contributes to the regulation of integrin activation and cell adhesion via the targeting of TLN1.


Asunto(s)
Regulación de la Expresión Génica , MicroARNs/metabolismo , Talina/metabolismo , Regiones no Traducidas 3' , Adhesión Celular , Línea Celular Tumoral , Membrana Celular/metabolismo , Biología Computacional , Epigénesis Genética , Células HCT116 , Células HEK293 , Humanos , Integrinas/metabolismo , Unión Proteica
7.
J Clin Med ; 10(18)2021 Sep 16.
Artículo en Inglés | MEDLINE | ID: mdl-34575296

RESUMEN

Background: A deregulated immune system has been implicated in the pathogenesis of post-cardiac arrest syndrome (PCAS). A soluble form of programmed cell death-1 (PD-1) ligand (sPD-L1) has been found at increased levels in cancer and sustained inflammation, thereby deregulating immune functions. Here, we aim to study the possible involvement of sPD-L1 in PCAS. Methods: Thirty out-of-hospital cardiac arrest (OHCA) patients consecutively admitted to the ER of Mie University Hospital were prospectively enrolled. Plasma concentrations of sPD-L1 were measured by an enzyme-linked immunosorbent assay in blood samples of all 30 OHCA patients obtained during cardiopulmonary resuscitation (CPR). In 13 patients who achieved return-of-spontaneous-circulation (ROSC), sPD-L1 levels were also measured daily in the ICU. Results: The plasma concentrations of sPD-L1 in OHCA were significantly increased; in fact, to levels as high as those observed in sepsis. sPD-L1 levels during CPR correlated with reduced peripheral lymphocyte counts and increased C-reactive protein levels. Of 13 ROSC patients, 7 cases survived in the ICU for more than 4 days. A longitudinal analysis of sPD-L1 levels in the 7 ROSC cases revealed that sPD-L1 levels occurred in parallel with organ failure. Conclusions: This study suggests that ischemia- reperfusion during CPR may aberrantly activate immune and endothelial cells to release sPD-L1 into circulation, which may play a role in the pathogenesis of immune exhaustion and organ failures associated with PCAS.

8.
J Gastroenterol Hepatol ; 36(11): 3149-3157, 2021 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-34370890

RESUMEN

BACKGROUND AND AIM: Epithelial regeneration, a critical step for the mucosal healing in inflammatory bowel disease, is tightly regulated by stem cells. Therefore, identification of the specific factors that induce stem cell proliferation could contribute to the development of effective strategies for treating inflammatory bowel disease. Recombinant soluble thrombomodulin (rsTM) has previously been shown to promote cell proliferation in skin and corneal wound healing in murine models, but its effects on intestinal epithelial cell proliferation remains unclear. METHODS: Mouse intestinal organoids and dextran sulfate sodium (DSS)-induced colitis mouse model were used to assess the effects of rsTM on proliferation of intestinal epithelial cells. The size and budding morphologies of organoids were studied by confocal microscopy. The gene expression levels were analyzed by quantitative real-time polymerase chain reaction and immunofluorescence analysis. The effects of rsTM on DSS-induced colitis were investigated by evaluating body weight changes, colon length, histological score, and survival rate. RESULTS: The rsTM markedly stimulated the growth of intestinal organoids, thereby increasing the surface areas and budding phenotypes of the organoids. rsTM also significantly upregulated the gene expression of intestinal stem cell-specific and epithelial cell-specific markers in a dose-dependent manner. Furthermore, the treatment with high concentrations of rsTM significantly improved the recovery of body weight, histological outcomes, colon length shortening, and prolonged the survival of mice with colitis. CONCLUSIONS: The rsTM promotes intestinal stem cell proliferation in intestinal organoids and enhances the mucosal healing during recovery phase in DSS-induced colitis.


Asunto(s)
Proliferación Celular , Colitis , Mucosa Intestinal , Trombomodulina , Animales , Proliferación Celular/fisiología , Colitis/inducido químicamente , Colitis/fisiopatología , Sulfato de Dextran/toxicidad , Modelos Animales de Enfermedad , Células Epiteliales/fisiología , Mucosa Intestinal/fisiología , Ratones , Ratones Endogámicos C57BL , Organoides/fisiología , Células Madre/fisiología , Trombomodulina/química , Trombomodulina/metabolismo , Cicatrización de Heridas
9.
Int J Mol Sci ; 22(15)2021 Aug 02.
Artículo en Inglés | MEDLINE | ID: mdl-34361061

RESUMEN

Sepsis is a sustained systemic inflammatory condition involving multiple organ failures caused by dysregulated immune response to infections. Sepsis induces substantial changes in energy demands at the cellular level leading to metabolic reprogramming in immune cells and stromal cells. Although sepsis-associated organ dysfunction and mortality have been partly attributed to the initial acute hyperinflammation and immunosuppression precipitated by a dysfunction in innate and adaptive immune responses, the late mortality due to metabolic dysfunction and immune paralysis currently represent the major problem in clinics. It is becoming increasingly recognized that intertissue and/or intercellular metabolic crosstalk via endocrine factors modulates maintenance of homeostasis, and pathological events in sepsis and other inflammatory diseases. Exosomes have emerged as a novel means of intercellular communication in the regulation of cellular metabolism, owing to their capacity to transfer bioactive payloads such as proteins, lipids, and nucleic acids to their target cells. Recent evidence demonstrates transfer of intact metabolic intermediates from cancer-associated fibroblasts via exosomes to modify metabolic signaling in recipient cells and promote cancer progression. Here, we review the metabolic regulation of endothelial cells and immune cells in sepsis and highlight the role of exosomes as mediators of cellular metabolic signaling in sepsis.


Asunto(s)
Células Endoteliales/patología , Exosomas/patología , Terapia de Inmunosupresión , Inflamación/patología , Enfermedades Metabólicas/patología , Sepsis/fisiopatología , Animales , Humanos , Inflamación/inmunología , Enfermedades Metabólicas/etiología
10.
Int J Mol Sci ; 22(7)2021 Mar 29.
Artículo en Inglés | MEDLINE | ID: mdl-33805523

RESUMEN

The intestinal epithelium serves as a dynamic barrier to protect the host tissue from exposure to a myriad of inflammatory stimuli in the luminal environment. Intestinal epithelial cells (IECs) encompass differentiated and specialized cell types that are equipped with regulatory genes, which allow for sensing of the luminal environment. Potential inflammatory cues can instruct IECs to undergo a diverse set of phenotypic alterations. Aging is a primary risk factor for a variety of diseases; it is now well-documented that aging itself reduces the barrier function and turnover of the intestinal epithelium, resulting in pathogen translocation and immune priming with increased systemic inflammation. In this study, we aimed to provide an effective epigenetic and regulatory outlook that examines age-associated alterations in the intestines through the profiling of microRNAs (miRNAs) on isolated mouse IECs. Our microarray analysis revealed that with aging, there is dysregulation of distinct clusters of miRNAs that was present to a greater degree in small IECs (22 miRNAs) compared to large IECs (three miRNAs). Further, miRNA-mRNA interaction network and pathway analyses indicated that aging differentially regulates key pathways between small IECs (e.g., toll-like receptor-related cascades) and large IECs (e.g., cell cycle, Notch signaling and small ubiquitin-related modifier pathway). Taken together, current findings suggest novel gene regulation pathways by epithelial miRNAs in aging within the gastrointestinal tissues.


Asunto(s)
Envejecimiento/fisiología , Células Epiteliales/fisiología , Mucosa Intestinal/citología , MicroARNs/fisiología , Animales , Simulación por Computador , Regulación de la Expresión Génica , Redes Reguladoras de Genes , Intestino Grueso/citología , Intestino Delgado/citología , Ratones Endogámicos C57BL , ARN Mensajero
11.
Int J Mol Sci ; 22(4)2021 Feb 13.
Artículo en Inglés | MEDLINE | ID: mdl-33668652

RESUMEN

Leukemia is a hematological malignancy that originates from hematopoietic stem cells in the bone marrow. Significant progress has made in understanding its pathogensis and in establishing chemotherapy and hematopoietic stem cell transplantation therapy (HSCT). However, while the successive development of new therapies, such as molecular-targeted therapy and immunotherapy, have resulted in remarkable advances, the fact remains that some patients still cannot be saved, and resistance to treatment and relapse are still problems that need to be solved in leukemia patients. The bone marrow (BM) niche is a microenvironment that includes hematopoietic stem cells and their supporting cells. Leukemia cells interact with bone marrow niches and modulate them, not only inducing molecular and functional changes but also switching to niches favored by leukemia cells. The latter are closely associated with leukemia progression, suppression of normal hematopoiesis, and chemotherapy resistance, which is precisely the area of ongoing study. Exosomes play an important role in cell-to-cell communication, not only with cells in close proximity but also with those more distant due to the nature of exosomal circulation via body fluids. In leukemia, exosomes play important roles in leukemogenesis, disease progression, and organ invasion, and their usefulness in the diagnosis and treatment of leukemia has recently been reported. The interaction between leukemia cell-derived exosomes and the BM microenvironment has received particular attention. Their interaction is believed to play a very important role; in addition to their diagnostic value, exosomes could serve as a marker for monitoring treatment efficacy and as an aid in overcoming drug resistance, among the many problems in leukemia patients that have yet to be overcome. In this paper, we will review bone marrow niches in leukemia, findings on leukemia-derived exosomes, and exosome-induced changes in bone marrow niches.


Asunto(s)
Médula Ósea/metabolismo , Comunicación Celular , Exosomas/metabolismo , Leucemia/metabolismo , Microambiente Tumoral , Médula Ósea/patología , Exosomas/patología , Humanos , Leucemia/patología
12.
Int J Mol Sci ; 22(4)2021 Feb 23.
Artículo en Inglés | MEDLINE | ID: mdl-33672100

RESUMEN

Integrins represent the biologically and medically significant family of cell adhesion molecules that govern a wide range of normal physiology. The activities of integrins in cells are dynamically controlled via activation-dependent conformational changes regulated by the balance of intracellular activators, such as talin and kindlin, and inactivators, such as Shank-associated RH domain interactor (SHARPIN) and integrin cytoplasmic domain-associated protein 1 (ICAP-1). The activities of integrins are alternatively controlled by homotypic lateral association with themselves to induce integrin clustering and/or by heterotypic lateral engagement with tetraspanin and syndecan in the same cells to modulate integrin adhesiveness. It has recently emerged that integrins are expressed not only in cells but also in exosomes, important entities of extracellular vesicles secreted from cells. Exosomal integrins have received considerable attention in recent years, and they are clearly involved in determining the tissue distribution of exosomes, forming premetastatic niches, supporting internalization of exosomes by target cells and mediating exosome-mediated transfer of the membrane proteins and associated kinases to target cells. A growing body of evidence shows that tumor and immune cell exosomes have the ability to alter endothelial characteristics (proliferation, migration) and gene expression, some of these effects being facilitated by vesicle-bound integrins. As endothelial metabolism is now thought to play a key role in tumor angiogenesis, we also discuss how tumor cells and their exosomes pleiotropically modulate endothelial functions in the tumor microenvironment.


Asunto(s)
Exosomas/metabolismo , Integrinas/metabolismo , Proteínas de la Membrana/metabolismo , Microambiente Tumoral/fisiología , Animales , Expresión Génica , Humanos , Integrinas/química , Neoplasias/metabolismo , Neoplasias/patología , Neovascularización Patológica/metabolismo , Proteínas del Tejido Nervioso/metabolismo , Talina/metabolismo
13.
Front Cell Dev Biol ; 9: 634853, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-33614663

RESUMEN

Extracellular vesicles (EVs) have emerged as key players of intercellular communication and mediate crosstalk between tissues. Metastatic tumors release tumorigenic EVs, capable of pre-conditioning distal sites for organotropic metastasis. Growing evidence identifies muscle cell-derived EVs and myokines as potent mediators of cellular differentiation, proliferation, and metabolism. Muscle-derived EVs cargo myokines and other biological modulators like microRNAs, cytokines, chemokines, and prostaglandins hence, are likely to modulate the remodeling of niches in vital sites, such as liver and adipose tissues. Despite the scarcity of evidence to support a direct relationship between muscle-EVs and cancer metastasis, their indirect attribution to the regulation of niche remodeling and the establishment of pre-metastatic homing niches can be put forward. This hypothesis is supported by the role of muscle-derived EVs in findings gathered from other pathologies like inflammation and metabolic disorders. In this review, we present and discuss studies that evidently support the potential roles of muscle-derived EVs in the events of niche pre-conditioning and remodeling of metastatic tumor microenvironment. We highlight the potential contributions of the integrin-mediated interactions with an emerging myokine, irisin, to the regulation of EV-driven microenvironment remodeling in tumor metastasis. Further research into muscle-derived EVs and myokines in cancer progression is imperative and may hold promising contributions to advance our knowledge in the pathophysiology, progression and therapeutic management of metastatic cancers.

14.
Biomedicines ; 9(2)2021 Feb 07.
Artículo en Inglés | MEDLINE | ID: mdl-33562346

RESUMEN

Thrombomodulin is a molecule with anti-coagulant and anti-inflammatory properties. Recently, thrombomodulin was reported to be able to bind extracellular matrix proteins, such as fibronectin and collagen; however, whether thrombomodulin regulates the binding of human breast cancer-derived cell lines to the extracellular matrix remains unknown. To investigate this, we created an extracellular domain of thrombomodulin, TMD123-Fc, or domain deletion TM-Fc proteins (TM domain 12-Fc, TM domain 23-Fc) and examined their bindings to fibronectin in vitro by ELISA. The lectin-like domain of thrombomodulin was found to be essential for the binding of the extracellular domain of thrombomodulin to fibronectin. Using a V-well cell adhesion assay or flow cytometry analysis with fluorescent beads, we found that both TMD123-Fc and TMD12-Fc inhibited the binding between ß1 integrin of human breast cancer-derived cell lines and fibronectin. Furthermore, TMD123-Fc and TMD12-Fc inhibited the binding of activated integrins to fibronectin under shear stress in the presence of Ca2+ and Mg2+ but not under strong integrin-activation conditions in the presence of Mg2+ without Ca2+. This suggests that thrombomodulin Fc fusion protein administered exogenously at a relatively early stage of inflammation may be applied to the development of new therapies that inhibit the binding of ß1 integrin of breast cancer cell lines to fibronectin.

15.
Methods Mol Biol ; 2217: 265-281, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-33215386

RESUMEN

Exosomes represent an important group of extracellular vesicles. They are formed in endosomal compartments and are actively secreted to extracellular spaces. Several membrane proteins, including integrins, are present on the surface of exosomes. As exosomal integrins are competent for binding to ligand, they can play important roles in directing the tissue distribution of exosomes. Integrin-directed exosomal trafficking in vivo is involved in regulating the remodeling of cell homing niches for metastatic cancers and migrating lymphocytes. This chapter describes the methods used to study integrin functions on exosomes including: isolation and biophysical characterization of exosomes, exosomal integrin-ligand binding assays, and in vivo competitive exosome homing assays.


Asunto(s)
Bioensayo , Exosomas/metabolismo , Integrinas/genética , Linfocitos/metabolismo , Traslado Adoptivo , Animales , Adhesión Celular , Línea Celular Tumoral , Centrifugación por Gradiente de Densidad , Endosomas/inmunología , Endosomas/metabolismo , Exosomas/inmunología , Exosomas/trasplante , Fluoresceínas/química , Colorantes Fluorescentes/química , Células HCT116 , Humanos , Integrinas/inmunología , Integrinas/metabolismo , Ligandos , Linfocitos/citología , Linfocitos/inmunología , Ratones , Unión Proteica , Transporte de Proteínas
16.
Front Cell Dev Biol ; 8: 588066, 2020.
Artículo en Inglés | MEDLINE | ID: mdl-33195249

RESUMEN

Integrins are transmembrane proteins that mediate cellular adhesion and migration to neighboring cells or the extracellular matrix, which is essential for cells to undertake diverse physiological and pathological pathways. For integrin activation and ligand binding, bidirectional signaling across the cell membrane is needed. Integrins aberrantly activated under pathologic conditions facilitate cellular infiltration into tissues, thereby causing inflammatory or tumorigenic progressions. Thus, integrins have emerged to the forefront as promising targets for developing therapeutics to treat autoimmune and cancer diseases. In contrast, it remains a fact that integrin-ligand interactions are beneficial for improving the health status of different tissues. Among these ligands, irisin, a myokine produced mainly by skeletal muscles in an exercise-dependent manner, has been shown to bind to integrin αVß5, alleviating symptoms under unfavorable conditions. These findings may provide insights into some of the underlying mechanisms by which exercise improves quality of life. This review will discuss the current understanding of integrin-ligand interactions in both health and disease. Likewise, we not only explain how diverse ligands play different roles in mediating cellular functions under both conditions via their interactions with integrins, but also specifically highlight the potential roles of the emerging ligand irisin in inflammation, cancer, and metabolic disease.

17.
Int J Mol Sci ; 21(22)2020 Nov 10.
Artículo en Inglés | MEDLINE | ID: mdl-33182773

RESUMEN

Sepsis is a systemic inflammatory disorder induced by a dysregulated immune response to infection resulting in dysfunction of multiple critical organs, including the intestines. Previous studies have reported contrasting results regarding the abilities of exosomes circulating in the blood of sepsis mice and patients to either promote or suppress inflammation. Little is known about how the gut epithelial cell-derived exosomes released in the intestinal luminal space during sepsis affect mucosal inflammation. To study this question, we isolated extracellular vesicles (EVs) from intestinal lavage of septic mice. The EVs expressed typical exosomal (CD63 and CD9) and epithelial (EpCAM) markers, which were further increased by sepsis. Moreover, septic-EV injection into inflamed gut induced a significant reduction in the messaging of pro-inflammatory cytokines TNF-a and IL-17A. MicroRNA (miRNA) profiling and reverse transcription and quantitative polymerase chain reaction (RT-qPCR) revealed a sepsis-induced exosomal increase in multiple miRNAs, which putatively target TNF-a and IL-17A. These results imply that intestinal epithelial cell (IEC)-derived luminal EVs carry miRNAs that mitigate pro-inflammatory responses. Taken together, our study proposes a novel mechanism by which IEC EVs released during sepsis transfer regulatory miRNAs to cells, possibly contributing to the amelioration of gut inflammation.


Asunto(s)
Interleucina-17/metabolismo , Mucosa Intestinal/inmunología , Sepsis/inmunología , Factor de Necrosis Tumoral alfa/metabolismo , Animales , Colitis/genética , Colitis/inmunología , Colitis/patología , Modelos Animales de Enfermedad , Exosomas/inmunología , Exosomas/patología , Vesículas Extracelulares/inmunología , Vesículas Extracelulares/patología , Humanos , Inflamación/genética , Inflamación/inmunología , Inflamación/patología , Interleucina-17/antagonistas & inhibidores , Interleucina-17/genética , Mucosa Intestinal/patología , Ratones , Ratones Endogámicos BALB C , MicroARNs/genética , MicroARNs/metabolismo , ARN Mensajero/genética , ARN Mensajero/metabolismo , Sepsis/genética , Sepsis/patología , Factor de Necrosis Tumoral alfa/antagonistas & inhibidores , Factor de Necrosis Tumoral alfa/genética
18.
Sci Immunol ; 5(52)2020 Oct 30.
Artículo en Inglés | MEDLINE | ID: mdl-33127608

RESUMEN

The presence of polyfunctional CD4+ T cells is often associated with favorable antitumor immunity. We report here that persistent activation of signal transducer and activator of transcription 5 (STAT5) in tumor-specific CD4+ T cells drives the development of polyfunctional T cells. We showed that ectopic expression of a constitutively active form of murine STAT5A (CASTAT5) enabled tumor-specific CD4+ T cells to undergo robust expansion, infiltrate tumors vigorously, and elicit antitumor CD8+ T cell responses in a CD4+ T cell adoptive transfer model system. Integrated epigenomic and transcriptomic analysis revealed that CASTAT5 induced genome-wide chromatin remodeling in CD4+ T cells and established a distinct epigenetic and transcriptional landscape. Single-cell RNA sequencing analysis further identified a subset of CASTAT5-transduced CD4+ T cells with a molecular signature indicative of progenitor polyfunctional T cells. The therapeutic significance of CASTAT5 came from our finding that adoptive transfer of T cells engineered to coexpress CD19-targeting chimeric antigen receptor (CAR) and CASTAT5 gave rise to polyfunctional CD4+ CAR T cells in a mouse B cell lymphoma model. The optimal therapeutic outcome was obtained when both CD4+ and CD8+ CAR T cells were transduced with CASTAT5, indicating that CASTAT5 facilitates productive CD4 help to CD8+ T cells. Furthermore, we provide evidence that CASTAT5 is functional in primary human CD4+ T cells, underscoring its potential clinical relevance. Our results implicate STAT5 as a valid candidate for T cell engineering to generate polyfunctional, exhaustion-resistant, and tumor-tropic antitumor CD4+ T cells to potentiate adoptive T cell therapy for cancer.


Asunto(s)
Linfocitos T CD4-Positivos/inmunología , Epigénesis Genética/inmunología , Inmunoterapia Adoptiva/métodos , Linfoma/terapia , Factor de Transcripción STAT5/metabolismo , Animales , Linfocitos T CD4-Positivos/metabolismo , Línea Celular Tumoral/trasplante , Modelos Animales de Enfermedad , Femenino , Regulación Neoplásica de la Expresión Génica/inmunología , Humanos , Linfoma/inmunología , Masculino , Ratones , Ratones Transgénicos , Cultivo Primario de Células , RNA-Seq , Receptores Quiméricos de Antígenos/inmunología , Factor de Transcripción STAT5/genética , Análisis de la Célula Individual , Transducción Genética
19.
Oncol Lett ; 20(4): 91, 2020 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-32831910

RESUMEN

Pyropia yezoensis Sookwawon 104 is a newly cultivated strain of red marine algae. The present study aimed to investigate the in vitro antiproliferative activity of sulfated polysaccharide extracted from P. yezoensis Sookwawon 104 (PYSP), as well as that of its low molecular weight (Mw) derivatives. PYSP is a heterogeneous sulfated polysaccharide mainly composed of galactose, glucose and fucose. PYSP was degraded by gamma-irradiation at doses of 20 and 100 kGy to produce two derivatives, named as PYSP-20 and PYSP-100, respectively. Comparison of PYSP, PYSP-20 and PYSP-100 revealed clear differences in their molecular weight (Mw) distributions, and distinct in vitro antiproliferative activities against Hep3B, MDA-MB-231 and HeLa cancer cell lines. PYSP-20 and PYSP-100 exhibited stronger antiproliferative effects than PYSP, suggesting that the reduction in Mw may have increased the in vitro antiproliferative activity. Furthermore, the mRNA expression levels of the antitumor gene P53 and cell cycle-associated genes P21, Cyclin B1 and cyclin dependent kinase 1 (Cdk1) were further analyzed by reverse transcription-quantitative PCR in PYSP-20 and PYSP-100-treated cancer cells. PYSP and its derivatives were shown to inhibit the proliferation of tumor cells by regulating the expression of P53, P21, Cyclin B1 and Cdk1. In conclusion, low-Mw polysaccharide derivatives prepared from P. yezoensis Sookwawon 104 by gamma-irradiation exhibit significant inhibition effects on cancer cell proliferation in vitro and may be a novel source of potential anticancer therapeutic agents.

20.
Diagn Pathol ; 15(1): 38, 2020 Apr 18.
Artículo en Inglés | MEDLINE | ID: mdl-32305065

RESUMEN

Exosomes represent an important subset of extracellular vesicles involved in inter-cellular communications in health and diseases. Exosomes secreted from cancer and immune cells travel to the specific tissues containing homing niches. The exosomes reaching the niches dynamically modify the gene expression and molecular architectures of the homing niche micro-environments. Cell adhesion molecule integrins regulate the tissue-specific homing patterns of not only cancer and immune cells, but also of the exosomes secreted from those cells. The exosome-mediated remodeling of the homing niches would affect immune lymphocyte migration and host defense, as well as cancer metastasis, thereby representing a potential therapeutic target.


Asunto(s)
Neoplasias de la Mama/inmunología , Neoplasias de la Mama/patología , Exosomas/inmunología , Exosomas/metabolismo , Integrinas/metabolismo , Animales , Femenino , Humanos , Integrinas/inmunología , Microambiente Tumoral/inmunología
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA