Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 13.949
Filtrar
1.
Nature ; 633(8028): 155-164, 2024 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-39232147

RESUMEN

Infectious, inflammatory and autoimmune conditions present differently in males and females. SARS-CoV-2 infection in naive males is associated with increased risk of death, whereas females are at increased risk of long COVID1, similar to observations in other infections2. Females respond more strongly to vaccines, and adverse reactions are more frequent3, like most autoimmune diseases4. Immunological sex differences stem from genetic, hormonal and behavioural factors5 but their relative importance is only partially understood6-8. In individuals assigned female sex at birth and undergoing gender-affirming testosterone therapy (trans men), hormone concentrations change markedly but the immunological consequences are poorly understood. Here we performed longitudinal systems-level analyses in 23 trans men and found that testosterone modulates a cross-regulated axis between type-I interferon and tumour necrosis factor. This is mediated by functional attenuation of type-I interferon responses in both plasmacytoid dendritic cells and monocytes. Conversely, testosterone potentiates monocyte responses leading to increased tumour necrosis factor, interleukin-6 and interleukin-15 production and downstream activation of nuclear factor kappa B-regulated genes and potentiation of interferon-γ responses, primarily in natural killer cells. These findings in trans men are corroborated by sex-divergent responses in public datasets and illustrate the dynamic regulation of human immunity by sex hormones, with implications for the health of individuals undergoing hormone therapy and our understanding of sex-divergent immune responses in cisgender individuals.


Asunto(s)
Testosterona , Personas Transgénero , Adulto , Femenino , Humanos , Masculino , Conjuntos de Datos como Asunto , Células Dendríticas/inmunología , Células Dendríticas/metabolismo , Células Dendríticas/efectos de los fármacos , Sistema Inmunológico/efectos de los fármacos , Sistema Inmunológico/metabolismo , Interferón Tipo I/inmunología , Interferón Tipo I/metabolismo , Interferón gamma/inmunología , Interferón gamma/metabolismo , Interleucina-15/inmunología , Interleucina-15/metabolismo , Células Asesinas Naturales/inmunología , Células Asesinas Naturales/efectos de los fármacos , Monocitos/inmunología , Monocitos/efectos de los fármacos , Monocitos/metabolismo , FN-kappa B/metabolismo , Caracteres Sexuales , Testosterona/efectos adversos , Testosterona/inmunología , Testosterona/farmacología , Testosterona/uso terapéutico , Factor de Necrosis Tumoral alfa/metabolismo
2.
Int J Biol Sci ; 20(11): 4114-4127, 2024.
Artículo en Inglés | MEDLINE | ID: mdl-39247831

RESUMEN

Osteosarcoma is the most prevalent form of primary malignant bone tumor, primarily affecting children and adolescents. The nerve growth factors (NGF) referred to as neurotrophins have been associated with cancer-induced bone pain; however, the role of NGF in osteosarcoma has yet to be elucidated. In osteosarcoma samples from the Genomic Data Commons data portal, we detected higher levels of NGF and M2 macrophage markers, but not M1 macrophage markers. In cellular experiments, NGF-stimulated osteosarcoma conditional medium was shown to facilitate macrophage polarization from the M0 to the M2 phenotype. NGF also enhanced VCAM-1-dependent monocyte adhesion within the osteosarcoma microenvironment by down-regulating miR-513c-5p levels through the FAK and c-Src cascades. In in vivo xenograft models, the overexpression of NGF was shown to enhance tumor growth, while the oral administration of the TrK inhibitor larotrectinib markedly antagonized NGF-promoted M2 macrophage expression and tumor progression. These results suggest that larotrectinib could potentially be used as a therapeutic agent aimed at mitigating NGF-mediated osteosarcoma progression.


Asunto(s)
Monocitos , Factor de Crecimiento Nervioso , Osteosarcoma , Microambiente Tumoral , Molécula 1 de Adhesión Celular Vascular , Osteosarcoma/metabolismo , Osteosarcoma/tratamiento farmacológico , Osteosarcoma/patología , Humanos , Factor de Crecimiento Nervioso/metabolismo , Animales , Microambiente Tumoral/efectos de los fármacos , Monocitos/metabolismo , Monocitos/efectos de los fármacos , Molécula 1 de Adhesión Celular Vascular/metabolismo , Ratones , Adhesión Celular/efectos de los fármacos , Línea Celular Tumoral , Neoplasias Óseas/metabolismo , Neoplasias Óseas/tratamiento farmacológico , Neoplasias Óseas/patología , Macrófagos/metabolismo , Pirimidinas/farmacología , Pirimidinas/uso terapéutico , Pirazoles/farmacología , Pirazoles/uso terapéutico , Ratones Desnudos
3.
Biomater Adv ; 165: 214024, 2024 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-39232353

RESUMEN

Graphene oxide (GO), a carbon-based nanomaterial, presents significant potential across biomedical fields such as bioimaging, drug delivery, biosensors, and phototherapy. This study examines the effects of integrating GO into poly(lactic-co-glycolic acid) (PLGA) scaffolds on human immune cell function. Our results demonstrate that high concentrations of GO reduce the viability of peripheral blood mononuclear cells (PBMCs) following stimulation with anti-CD3 antibody. This reduction extends to T lymphocyte activation, evident from the diminished proliferative response to T cell receptor engagement and impaired differentiation into T helper subsets and regulatory T cells. Interestingly, although GO induces a minimal response in resting monocytes, but it significantly affects both the viability and the differentiation potential of monocytes induced to mature toward M1 pro-inflammatory and M2-like immunoregulatory macrophages. This study seeks to address a critical gap by investigating the in vitro immunomodulatory effects of PLGA scaffolds incorporating various concentrations of GO on primary immune cells, specifically PBMCs isolated from healthy donors. Our findings emphasize the need to optimize the GO to PLGA ratios and scaffold design to advance PLGA-GO-based biomedical applications. STATEMENT OF SIGNIFICANCE: Graphene oxide (GO) holds immense promise for biomedical applications due to its unique properties. However, concerns regarding its potential to trigger adverse immune responses remain. This study addresses this critical gap by investigating the in vitro immunomodulatory effects of PLGA scaffolds incorporating increasing GO concentrations on human peripheral blood mononuclear cells (PBMCs). By elucidating the impact on cell viability, T cell proliferation and differentiation, and the maturation/polarization of antigen-presenting cells, this work offers valuable insights for designing safe and immunologically compatible GO-based biomaterials for future clinical translation.


Asunto(s)
Grafito , Leucocitos Mononucleares , Copolímero de Ácido Poliláctico-Ácido Poliglicólico , Andamios del Tejido , Grafito/química , Grafito/farmacología , Humanos , Copolímero de Ácido Poliláctico-Ácido Poliglicólico/química , Copolímero de Ácido Poliláctico-Ácido Poliglicólico/farmacología , Leucocitos Mononucleares/efectos de los fármacos , Leucocitos Mononucleares/inmunología , Andamios del Tejido/química , Diferenciación Celular/efectos de los fármacos , Supervivencia Celular/efectos de los fármacos , Proliferación Celular/efectos de los fármacos , Activación de Linfocitos/efectos de los fármacos , Materiales Biocompatibles/farmacología , Materiales Biocompatibles/química , Monocitos/efectos de los fármacos , Monocitos/inmunología , Macrófagos/efectos de los fármacos , Macrófagos/inmunología
4.
Cell Biochem Funct ; 42(7): e4115, 2024 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-39264203

RESUMEN

In this study, the protective effects of Panax notoginseng saponins (PNS) against gamma radiation-induced DNA damage and associated physiological alterations in Swiss albino mice were investigated. Exposure to gamma radiation led to a dose-dependent increase in cytokinesis-blocked micronuclei (CBMN) double-strand DNA breaks (DSBs), dicentric aberrations (DC), formation in peripheral blood mononuclear cells. However, pretreatment with PNS at concentrations of 1, 5, and 10 µg/mL significantly attenuated the frequencies of DC and CBMN in a concentration-dependent manner. PNS administration before radiation exposure also reduced radiation-induced DSBs in BL, indicating protection against reactive oxygen species generation and DNA damage. Notably, pretreatment with PNS at 10 µg/mL prevented the overexpression of γ-H2AX, proteins associated with DNA damage response, in irradiated mice. In addition, in vivo studies showed intraperitoneal administration of PNS (25 mg/kg body weight) for 1 h before radiation exposure mitigated lipid peroxidation levels and restored antioxidant status, countering oxidative damage induced by gamma radiation. Furthermore, PNS pretreatment reversed the decrease in hemoglobin (Hb) content, white blood cell count, and red blood cell count in irradiated mice, indicating preservation of hematological parameters. Overall, PNS demonstrated an anticlastogenic effect by modulating radiation-induced DSBs and preventing oxidative damage, thus highlighting its potential as a protective agent against radiation-induced DNA damage and associated physiological alterations. Clinically, PNS will be beneficial for cancer patients undergoing radiotherapy, but their pharmacological properties and toxicity profiles need to be studied.


Asunto(s)
Rayos gamma , Panax notoginseng , Saponinas , Animales , Rayos gamma/efectos adversos , Saponinas/farmacología , Ratones , Panax notoginseng/química , Humanos , Masculino , Daño del ADN/efectos de los fármacos , Leucocitos Mononucleares/efectos de los fármacos , Leucocitos Mononucleares/metabolismo , Leucocitos Mononucleares/efectos de la radiación , Estrés Oxidativo/efectos de los fármacos , Estrés Oxidativo/efectos de la radiación , Protectores contra Radiación/farmacología , Roturas del ADN de Doble Cadena/efectos de los fármacos , Roturas del ADN de Doble Cadena/efectos de la radiación , Monocitos/efectos de los fármacos , Monocitos/metabolismo , Monocitos/efectos de la radiación , Especies Reactivas de Oxígeno/metabolismo , Antioxidantes/farmacología
5.
Immunohorizons ; 8(9): 668-678, 2024 Sep 01.
Artículo en Inglés | MEDLINE | ID: mdl-39259207

RESUMEN

Systemic lupus erythematosus is an autoimmune disease characterized by excessive inflammation and production of pathogenic Abs. Histone deacetylase 6 (HDAC6) is a class IIb histone deacetylase. It has been reported that selective HDAC6 inhibition decreases inflammation in lupus mouse models. In this study, sex- and age-matched wild-type (WT) and HDAC6-/- mice on the C57BL/6 background were administered 0.5 ml of pristane or PBS i.p. at 8-12 wk of age and were euthanized 10 d later. At sacrifice, body weight and spleen weight were measured, sera were collected, and splenocytes and peritoneal cells were harvested for flow cytometry. We found pristane administration increased the spleen weight with no difference between WT and HDAC6-/- mice. Pristane administration promoted the population of CD11b+Ly6C++ inflammatory monocytes and CD11b+Ly6G+ neutrophils. Peritoneal recruitment of these inflammatory monocytes and neutrophils was significantly decreased in HDAC6-/- mice compared with the WT mice. Flow cytometry results showed that the number of CD69+ T and B cells was increased in HDAC6-/- mice. Pristane administration also induced the IFN signature genes as determined by RT-qPCR. Furthermore, IFN signature genes were not affected in HDAC6-/- mice compared with the WT mice. In vitro studies in J774A.1 cells revealed that the selective HDAC6 inhibitor (ACY-738) increased acetylation of NF-κB while increasing Stat1 phosphorylation, which resulted in inducible NO synthase production in LPS/IFN-γ-stimulated cells. Taken together, these results demonstrate that although HDAC6 inhibition may inhibit some inflammatory pathways, others remain unaffected.


Asunto(s)
Histona Desacetilasa 6 , Inflamación , Ratones Endogámicos C57BL , Ratones Noqueados , Terpenos , Animales , Histona Desacetilasa 6/antagonistas & inhibidores , Histona Desacetilasa 6/genética , Histona Desacetilasa 6/metabolismo , Terpenos/farmacología , Ratones , Lupus Eritematoso Sistémico/genética , Lupus Eritematoso Sistémico/inmunología , Bazo/inmunología , Bazo/metabolismo , Bazo/citología , Monocitos/metabolismo , Monocitos/inmunología , Monocitos/efectos de los fármacos , Femenino , Modelos Animales de Enfermedad , Neutrófilos/inmunología , Neutrófilos/metabolismo , Factor de Transcripción STAT1/metabolismo , Masculino , Linfocitos B/inmunología , Linfocitos B/metabolismo
6.
J Autoimmun ; 148: 103300, 2024 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-39116634

RESUMEN

The mechanisms of endotoxin tolerance (ET), which down-regulate inflammation, are well described in response to exogenous toll-like receptor ligands, but few studies have focused on ET-associated mechanisms in inflammatory disease. As blocking TNF can attenuate the development of ET, the effect of anti-TNF on the expression of key ET-associated molecules in inflammatory auto-immune disease was measured; changes in inflammatory gene expression were confirmed using an ET bioassay. The expression of immunomodulatory molecules was measured in a murine model of arthritis treated with anti-TNF and the expression of ET-associated molecules was measured in whole blood in rheumatoid arthritis (RA) and ankylosing spondylitis (AS) patients, before and after therapy. The expression of ET-associated genes was also measured in RA patient monocytes before and after therapy, in anti-TNF responders and non-responders. Tnfaip3, Ptpn6 and Irak3 were differentially expressed in affected paws, spleens, lymph nodes and circulating leucocytes in experimental murine arthritis treated with anti-TNF. Prior to therapy, the expression of TNFAIP3, INPP5D, PTPN6, CD38 and SIGIRR in whole blood differed between human healthy controls and RA or AS patients. In blood monocytes from RA patients, the expression of TNFAIP3 was significantly reduced by anti-TNF therapy in non-responders. Prior to therapy, anti-TNF non-responders had higher expression of TNFAIP3 and SLPI, compared to responders. Although the expression of TNFAIP3 was significantly higher in RA non-responders prior to treatment, the post-treatment reduction to a level similar to responders did not coincide with a clinical response to therapy.


Asunto(s)
Artritis Reumatoide , Endotoxinas , Tolerancia Inmunológica , Espondilitis Anquilosante , Proteína 3 Inducida por el Factor de Necrosis Tumoral alfa , Factor de Necrosis Tumoral alfa , Animales , Humanos , Ratones , Artritis Reumatoide/tratamiento farmacológico , Artritis Reumatoide/inmunología , Tolerancia Inmunológica/efectos de los fármacos , Proteína 3 Inducida por el Factor de Necrosis Tumoral alfa/metabolismo , Proteína 3 Inducida por el Factor de Necrosis Tumoral alfa/genética , Endotoxinas/inmunología , Espondilitis Anquilosante/tratamiento farmacológico , Espondilitis Anquilosante/inmunología , Factor de Necrosis Tumoral alfa/metabolismo , Factor de Necrosis Tumoral alfa/antagonistas & inhibidores , Péptidos y Proteínas de Señalización Intracelular/genética , Péptidos y Proteínas de Señalización Intracelular/metabolismo , Masculino , Quinasas Asociadas a Receptores de Interleucina-1/metabolismo , Quinasas Asociadas a Receptores de Interleucina-1/genética , Femenino , Proteína Tirosina Fosfatasa no Receptora Tipo 6/metabolismo , Artritis Experimental/inmunología , Artritis Experimental/tratamiento farmacológico , Proteínas de Unión al ADN/metabolismo , Proteínas de Unión al ADN/genética , Monocitos/inmunología , Monocitos/metabolismo , Monocitos/efectos de los fármacos , Persona de Mediana Edad , Adulto , Inflamación/inmunología , Modelos Animales de Enfermedad
7.
Int J Mol Sci ; 25(16)2024 Aug 06.
Artículo en Inglés | MEDLINE | ID: mdl-39201246

RESUMEN

As a nutraceutical, bovine lactoferrin (bLf), an iron-binding glycoprotein involved in innate immunity, is gaining elevated attention for its ability to exert pleiotropic functions and to be exceptionally tolerated even at high dosages. Some of bLf's activities, including its anti-inflammatory and antioxidant, are tightly linked to its ability to both chelate iron and enter inside the cell nucleus. Here, we present data about Valpalf®, a new formulation containing bLf, sodium citrate, and sodium bicarbonate at a molar ratio of 10-3. In the present study, Valpalf® exhibits superior iron-binding capacity, resistance to tryptic digestion, and a greater capacity to accumulate into the nucleus over time when compared to the native bLf alone. In agreement, Valpalf® effectively reduces interleukin(IL)-6 levels in lipopolysaccharide-stimulated macrophages and modulates the expression of antioxidant enzymes, such as superoxide dismutase 1 and 2, in phorbol-12-myristate-13-acetate-stimulated monocytes. Of note, this potentiated bioactivity was corroborated in a retrospective study on the treatment of anemia of inflammation in hereditary thrombophilic pregnant and non-pregnant women, demonstrating that Valpalf® improves hematological parameters and reduces serum IL-6 levels to a higher extent than bLf alone.


Asunto(s)
Suplementos Dietéticos , Interleucina-6 , Lactoferrina , Superóxido Dismutasa , Lactoferrina/farmacología , Lactoferrina/química , Animales , Bovinos , Humanos , Femenino , Superóxido Dismutasa/metabolismo , Interleucina-6/metabolismo , Antioxidantes/farmacología , Antioxidantes/química , Ratones , Citrato de Sodio/farmacología , Superóxido Dismutasa-1/metabolismo , Bicarbonato de Sodio/farmacología , Bicarbonato de Sodio/química , Embarazo , Macrófagos/efectos de los fármacos , Macrófagos/metabolismo , Antiinflamatorios/farmacología , Antiinflamatorios/química , Monocitos/efectos de los fármacos , Monocitos/metabolismo , Hierro/metabolismo , Lipopolisacáridos , Anemia/tratamiento farmacológico
8.
Cells ; 13(15)2024 Aug 04.
Artículo en Inglés | MEDLINE | ID: mdl-39120331

RESUMEN

Hypertension (HTN) impacts almost half of adults, predisposing them to cardiovascular disease and renal damage. Salt-sensitive HTN (SSHTN) and angiotensin II (A2)-induced HTN (A2HTN) both involve immune system activation and renal innate immune cell infiltration. Subpopulations of activated [Cluster of differentiation 38 (CD38)] innate immune cells, such as macrophages and dendritic cells (DCs), play distinct roles in modulating renal function and blood pressure. It is unknown how these cells become CD38+ or which subtypes are pro-hypertensive. When bone marrow-derived monocytes (BMDMs) were grown in granulocyte-macrophage colony stimulating factor (GM-CSF) and treated with salt or A2, CD38+ macrophages and CD38+ DCs increased. The adoptive transfer of GM-CSF-primed BMDMs into mice with either SSHTN or A2HTN increased renal CD38+ macrophages and CD38+ DCs. Flow cytometry revealed increased renal M1 macrophages and type-2 conventional DCs (cDC2s), along with their CD38+ counterparts, in mice with either SSHTN or A2HTN. These results were replicable in vitro. Either salt or A2 treatment of GM-CSF-primed BMDMs significantly increased bone marrow-derived (BMD)-M1 macrophages, CD38+ BMD-M1 macrophages, BMD-cDC2s, and CD38+ BMD-cDC2s. Overall, these data suggest that GM-CSF is necessary for the salt or A2 induction of CD38+ innate immune cells, and that CD38 distinguishes pro-hypertensive immune cells. Further investigation of CD38+ M1 macrophages and CD38+ cDC2s could provide new therapeutic targets for both SSHTN and A2HTN.


Asunto(s)
Angiotensina II , Células Dendríticas , Factor Estimulante de Colonias de Granulocitos y Macrófagos , Inmunidad Innata , Macrófagos , Animales , Angiotensina II/farmacología , Factor Estimulante de Colonias de Granulocitos y Macrófagos/farmacología , Factor Estimulante de Colonias de Granulocitos y Macrófagos/metabolismo , Ratones , Inmunidad Innata/efectos de los fármacos , Macrófagos/efectos de los fármacos , Macrófagos/inmunología , Macrófagos/metabolismo , Células Dendríticas/efectos de los fármacos , Células Dendríticas/inmunología , Células Dendríticas/metabolismo , Hipertensión/inmunología , Ratones Endogámicos C57BL , ADP-Ribosil Ciclasa 1/metabolismo , Masculino , Monocitos/efectos de los fármacos , Monocitos/metabolismo , Monocitos/inmunología , Riñón/inmunología , Riñón/efectos de los fármacos
9.
Food Chem Toxicol ; 192: 114912, 2024 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-39121895

RESUMEN

Food grade titanium dioxide E171 has been used in products such as confectionery, doughs and flours to enhance organoleptic properties. The European Union has warned about adverse effects on humans due to oral consumption. After oral exposure, E171 reaches the bloodstream which raises the concern about effects on blood cells such as monocytes. One of the main functions of these cells is the differentiation of macrophages leading to the phagocytosis of foreign particles. The aim of this study was to evaluate the effect of E171 exposure on the phagocytic capacity and differentiation process of monocytes (THP-1) into macrophages. Physicochemical E171 properties were evaluated, and THP-1 monocytes were exposed to 4, 40 and 200 µg/ml. Cell viability, uptake capacity, cytokine release, the differentiation process, cytoskeletal arrangement and E171 internalization were assayed. Results showed that E171 particles had an amorphous shape with a mean of hydrodynamic size of ∼46 nm in cell culture media. Cell viability decreased until the 9th day of exposure, while the uptake capacity decreased up to 62% in a concentration dependent manner in monocytes. Additionally, the E171 exposure increased the proinflammatory cytokines release and decreased the cell differentiation by a 61% in macrophages. E171 induced changes in cytoskeletal arrangement and some of the E171 particles were located inside the nuclei. We conclude that E171 exposure in THP-1 monocytes induced an inflammatory response, impaired the phagocytic capacity, and interfered with cell differentiation from monocytes to macrophages.


Asunto(s)
Diferenciación Celular , Supervivencia Celular , Macrófagos , Monocitos , Fagocitosis , Titanio , Titanio/toxicidad , Titanio/química , Humanos , Diferenciación Celular/efectos de los fármacos , Fagocitosis/efectos de los fármacos , Macrófagos/efectos de los fármacos , Monocitos/efectos de los fármacos , Supervivencia Celular/efectos de los fármacos , Citocinas/metabolismo , Células THP-1
10.
Exp Cell Res ; 442(1): 114216, 2024 Sep 01.
Artículo en Inglés | MEDLINE | ID: mdl-39182663

RESUMEN

The toxic effect of oxidized-heme, also known as hemin, is implicated in developing adverse clinical outcome in various hematolytic diseases. To simulate and reconstruct the molecular events associated with hemin exposure on circulating monocytes, we employed a THP-1 cell line based in vitro model. Flow cytometry and Western blot analyses were subsequently applied. Hemin-treated THP-1 produced ROS in a dose-dependent manner which resulted in 10-30 % of cell death primarily through apoptosis. Surviving cells induced autophagy which too was ROS-dependent, as revealed by application of N-acetyl-L-cysteine. Hemin-mediated autophagy promoted differentiation of CD14+ THP-1 cells into CD11b+ macrophages. Application of 3-methyladenine, reinforced that differentiation of THP-1 was an autophagy-dependent process. It was revealed that despite a higher polarization towards M2-macrophage, synthesis of pro-inflammatory cytokines namely TNF-α, IL-1A, IL-2, IL-8 and IL-17A predominated. IL-6, a pleiotropic cytokine, was also elevated. It may thus be surmised that hemin-induced pro-inflammatory response in THP-1 is downstream to ROS-dependent autophagy and monocyte differentiation. This finding is translationally meaningful as hemin is already approved by FDA for amelioration of acute porphyria and is actively considered as a therapeutic agent for other diseases. This study underscores the need of further research untangling the reciprocal regulation of inflammatory signaling and autophagy under oxidative stress.


Asunto(s)
Autofagia , Diferenciación Celular , Hemina , Macrófagos , Especies Reactivas de Oxígeno , Humanos , Especies Reactivas de Oxígeno/metabolismo , Autofagia/efectos de los fármacos , Diferenciación Celular/efectos de los fármacos , Hemina/farmacología , Macrófagos/metabolismo , Macrófagos/efectos de los fármacos , Células THP-1 , Inflamación/metabolismo , Inflamación/patología , Citocinas/metabolismo , Monocitos/metabolismo , Monocitos/efectos de los fármacos , Apoptosis/efectos de los fármacos
11.
Cancer Med ; 13(16): e70065, 2024 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-39190576

RESUMEN

AIM: Lenvatinib mesylate (LEN) is an oral tyrosine kinase inhibitor used to treat various cancers, including hepatocellular carcinoma (HCC). HCC treatment with LEN is associated with a very high incidence of adverse events. This study was aimed at investigating the incidence of LEN-induced palmar-planter erythrodysesthesia syndrome (PPES) and its relationship with patient demographics by analyzing clinical laboratory data of LEN-treated patients with HCC. METHODS: This was a single-centre, retrospective study of patients with HCC who received LEN between April 19, 2018, and September 30, 2020. The observation period was from 1 week before the start of LEN administration to 1 month after the end of administration. RESULTS: Overall, 75 patients with HCC were enrolled. LEN-induced PPES was found in 48.0% (36/75 patients). In these patients, alkaline phosphatase (ALP), γ-Glutamyl transpeptidase (γ-GTP) and monocytes (MONO) were significantly high (ALP: p = 1.32 × 10-3, γ-GTP: p = 4.25 × 10-3 and MONO: p = 0.013). The cut off values of ALP, γ-GTP and MONO for LEN-induced PPES were estimated at 573 U/L, 89 U/L, and 310 counts/µL, respectively. In the multivariate analysis, γ-GTP and MONO were independent risk factors for LEN-induced PPES. CONCLUSIONS: High γ-GTP and high MONO were risk factors for LEN-induced PPES.


Asunto(s)
Carcinoma Hepatocelular , Síndrome Mano-Pie , Neoplasias Hepáticas , Compuestos de Fenilurea , Quinolinas , Humanos , Carcinoma Hepatocelular/tratamiento farmacológico , Masculino , Neoplasias Hepáticas/tratamiento farmacológico , Estudios Retrospectivos , Femenino , Anciano , Quinolinas/efectos adversos , Quinolinas/uso terapéutico , Persona de Mediana Edad , Factores de Riesgo , Compuestos de Fenilurea/efectos adversos , Compuestos de Fenilurea/administración & dosificación , Síndrome Mano-Pie/etiología , Síndrome Mano-Pie/epidemiología , gamma-Glutamiltransferasa/sangre , Inhibidores de Proteínas Quinasas/efectos adversos , Inhibidores de Proteínas Quinasas/uso terapéutico , Fosfatasa Alcalina/sangre , Anciano de 80 o más Años , Antineoplásicos/efectos adversos , Incidencia , Monocitos/efectos de los fármacos , Adulto
12.
J Pharmacol Sci ; 156(2): 125-133, 2024 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-39179331

RESUMEN

Benproperine (BNP) is a nonnarcotic antitussive drug that is used to treat bronchitis. In the present study, we examined the anti-inflammatory effects of BNP in vitro and in vivo. BNP was found to reduce the secretion of pro-inflammatory cytokines, such as interleukin (IL)-6, in lipopolysaccharide (LPS)-treated RAW264.7 monocyte/macrophage-lineage cells in vitro. As IL-6 is a biomarker for sepsis and has been suggested to exacerbate symptoms, we used an animal model to determine whether BNP reduces IL-6 levels in vivo and improves sepsis symptoms. Notably, BNP reduced IL-6 levels in the lungs of LPS-treated mice and improved LPS-induced hypothermia, one of the symptoms of sepsis. BNP reduced the mortality of septic mice administered a lethal dose of LPS. To reveal the mechanisms underlying the anti-inflammatory function of BNP, we assessed intracellular signaling in LPS-treated RAW264.7 cells. BNP induced the phosphorylation of protein kinase B (Akt) in RAW264.7 cells with/without LPS treatment. Wortmannin, an inhibitor of phosphoinositide 3-kinase reduced the phosphorylation levels of Akt. Wortmannin also obstructed the reduction of IL-6 secretion caused by BNP. Altogether, BNP was found to exhibit an anti-inflammatory function via Akt signaling. Therefore, BNP could be a drug candidate for inflammatory diseases, including sepsis.


Asunto(s)
Modelos Animales de Enfermedad , Interleucina-6 , Lipopolisacáridos , Macrófagos , Monocitos , Proteínas Proto-Oncogénicas c-akt , Sepsis , Transducción de Señal , Animales , Ratones , Sepsis/tratamiento farmacológico , Sepsis/metabolismo , Interleucina-6/metabolismo , Proteínas Proto-Oncogénicas c-akt/metabolismo , Transducción de Señal/efectos de los fármacos , Macrófagos/metabolismo , Macrófagos/efectos de los fármacos , Células RAW 264.7 , Masculino , Monocitos/metabolismo , Monocitos/efectos de los fármacos , Antiinflamatorios/farmacología , Fosforilación/efectos de los fármacos , Ratones Endogámicos C57BL , Fosfatidilinositol 3-Quinasas/metabolismo
13.
Front Immunol ; 15: 1403263, 2024.
Artículo en Inglés | MEDLINE | ID: mdl-39086490

RESUMEN

Background: Cell energy metabolism controls the activation and function of dendritic cells (DCs). Inflammatory dendritic epidermal cells (IDECs) in skin lesions of atopic dermatitis (AD) express high-affinity IgE receptor (FcϵRI) and toll-like receptor 2 (TLR2), which mediate the generation and maintenance of inflammation. However, cellular energy metabolism and effector function of IDECs mediated by FcϵRI and TLR2 have not been fully elucidated. Methods: IDECs in vitro were treated with TLR2 agonist Pam3CSK4 and anti-IgE alone or in combination for 24 h. Further, we analyzed the expression of cell surface activation markers, production of inflammatory factors, and cellular energy metabolism profiles of IDECs by using flow cytometry, multiplex assay, RNA sequencing, targeted energy metabolism, and seahorse assays. Results: Compared to the unstimulated or anti-IgE groups, Pam3CSK4 alone or combined with anti-IgE groups significantly increased the expression of CD80, CD83, and CD86 on IDECs, but did not affect the expression of the above markers in the anti-IgE group. The release of inflammatory cytokines increased in the Pam3CSK4 alone or combined with anti-IgE groups, while there was a weak increasing trend in the anti-IgE group. The glycolysis/gluconeogenesis pathway of carbon metabolism was affected in all treatment groups. Furthermore, compared to the control group, we found a decrease in pyruvic acid, upregulation of PFKM, downregulation of FBP1, and increase in extracellular lactate, glycolysis rate, and glycolysis capacity after all treatments, while there was no difference between each treatment group. However, there was no difference in glycolytic reserve and mitochondrial basic and maximum respiration among all groups. Conclusion: Our results indicate that glycolysis of IDECs may be activated through FcϵRI and TLR2 to upregulate inflammatory factors, suggesting that danger signals from bacteria or allergens might evoke an inflammatory response from AD through the glycolysis pathway.


Asunto(s)
Células Dendríticas , Glucosa , Lipopéptidos , Monocitos , Receptor Toll-Like 2 , Humanos , Lipopéptidos/farmacología , Células Dendríticas/inmunología , Células Dendríticas/metabolismo , Células Dendríticas/efectos de los fármacos , Monocitos/inmunología , Monocitos/metabolismo , Monocitos/efectos de los fármacos , Glucosa/metabolismo , Receptor Toll-Like 2/metabolismo , Receptor Toll-Like 2/agonistas , Dermatitis Atópica/inmunología , Dermatitis Atópica/metabolismo , Metabolismo Energético/efectos de los fármacos , Inflamación/inmunología , Inflamación/metabolismo , Células Cultivadas , Receptores de IgE/metabolismo , Citocinas/metabolismo , Inmunoglobulina E/inmunología , Glucólisis , Diferenciación Celular
14.
EMBO Rep ; 25(8): 3276-3299, 2024 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-39039298

RESUMEN

Transmigration of circulating monocytes from the bloodstream to tissues represents an early hallmark of inflammation. This process plays a pivotal role during viral neuroinvasion, encephalitis, and HIV-associated neurocognitive disorders. How monocytes locally unzip endothelial tight junction-associated proteins (TJAPs), without perturbing impermeability, to reach the central nervous system remains poorly understood. Here, we show that human circulating monocytes express the TJAP Occludin (OCLN) to promote transmigration through endothelial cells. We found that human monocytic OCLN (hmOCLN) clusters at monocyte-endothelium interface, while modulation of hmOCLN expression significantly impacts monocyte transmigration. Furthermore, we designed OCLN-derived peptides targeting its extracellular loops (EL) and show that transmigration of treated monocytes is inhibited in vitro and in zebrafish embryos, while preserving vascular integrity. Monocyte transmigration toward the brain is an important process for HIV neuroinvasion and we found that the OCLN-derived peptides significantly inhibit HIV dissemination to cerebral organoids. In conclusion, our study identifies an important role for monocytic OCLN during transmigration and provides a proof-of-concept for the development of mitigation strategies to prevent monocyte infiltration and viral neuroinvasion.


Asunto(s)
Células Endoteliales , Monocitos , Ocludina , Migración Transendotelial y Transepitelial , Pez Cebra , Ocludina/metabolismo , Ocludina/genética , Humanos , Monocitos/metabolismo , Monocitos/efectos de los fármacos , Monocitos/virología , Animales , Migración Transendotelial y Transepitelial/efectos de los fármacos , Células Endoteliales/metabolismo , Células Endoteliales/virología , Células Endoteliales/efectos de los fármacos , Infecciones por VIH/virología , Infecciones por VIH/tratamiento farmacológico , VIH-1/fisiología , VIH-1/efectos de los fármacos , Péptidos/farmacología , Péptidos/metabolismo , Encéfalo/metabolismo , Encéfalo/virología
15.
J Immunol Methods ; 532: 113716, 2024 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-38960065

RESUMEN

The human monocytic THP-1 cell line is the most routinely employed in vitro model for studying monocyte-to-macrophage differentiation. Despite the wide use of this model, differentiation protocols using phorbol 12-myristate-13-acetate (PMA) or 1,25-dihydroxyvitamin D3 (1,25D3) vary drastically between studies. Given that differences in differentiation protocols have the potential to impact the characteristics of the macrophages produced, we aimed to assess the efficacy of three different THP-1 differentiation protocols by assessing changes in morphology and gene- and cell surface macrophage marker expression. THP-1 cells were differentiated with either 5 nM PMA, 10 nM 1,25D3, or a combination thereof, followed by a rest period. The results indicated that all three protocols significantly increased the expression of the macrophage markers, CD11b (p < 0.001) and CD14 (p < 0.010). Despite this, THP-1 cells exposed to 1,25D3 alone did not adopt the morphological and expression characteristics associated with macrophages. PMA was required to produce these characteristics, which were found to be more pronounced in the presence of 1,25D3. Both PMA- and PMA with 1,25D3-differentiated THP-1 cells were capable of M1 and M2 macrophage polarization, though the gene expression of polarization-associated markers was most pronounced in PMA with 1,25D3-differentiated THP-1 cells. Moreover, the combination of PMA with 1,25D3 appeared to support the process of commitment to a particular polarization state.


Asunto(s)
Calcitriol , Diferenciación Celular , Macrófagos , Monocitos , Acetato de Tetradecanoilforbol , Humanos , Diferenciación Celular/efectos de los fármacos , Acetato de Tetradecanoilforbol/farmacología , Macrófagos/efectos de los fármacos , Macrófagos/metabolismo , Células THP-1 , Monocitos/efectos de los fármacos , Monocitos/metabolismo , Monocitos/citología , Calcitriol/farmacología , Receptores de Lipopolisacáridos/metabolismo , Antígeno CD11b/metabolismo
16.
Cell ; 187(16): 4318-4335.e20, 2024 Aug 08.
Artículo en Inglés | MEDLINE | ID: mdl-38964327

RESUMEN

Dexamethasone is a life-saving treatment for severe COVID-19, yet its mechanism of action is unknown, and many patients deteriorate or die despite timely treatment initiation. Here, we identify dexamethasone treatment-induced cellular and molecular changes associated with improved survival in COVID-19 patients. We observed a reversal of transcriptional hallmark signatures in monocytes associated with severe COVID-19 and the induction of a monocyte substate characterized by the expression of glucocorticoid-response genes. These molecular responses to dexamethasone were detected in circulating and pulmonary monocytes, and they were directly linked to survival. Monocyte single-cell RNA sequencing (scRNA-seq)-derived signatures were enriched in whole blood transcriptomes of patients with fatal outcome in two independent cohorts, highlighting the potential for identifying non-responders refractory to dexamethasone. Our findings link the effects of dexamethasone to specific immunomodulation and reversal of monocyte dysregulation, and they highlight the potential of single-cell omics for monitoring in vivo target engagement of immunomodulatory drugs and for patient stratification for precision medicine approaches.


Asunto(s)
Tratamiento Farmacológico de COVID-19 , COVID-19 , Dexametasona , Monocitos , SARS-CoV-2 , Análisis de la Célula Individual , Humanos , Dexametasona/farmacología , Dexametasona/uso terapéutico , Monocitos/metabolismo , Monocitos/efectos de los fármacos , SARS-CoV-2/efectos de los fármacos , Masculino , Femenino , Transcriptoma , Persona de Mediana Edad , Anciano , Glucocorticoides/uso terapéutico , Glucocorticoides/farmacología , Pulmón/patología , Adulto
17.
Aging (Albany NY) ; 16(14): 11134-11150, 2024 Jul 26.
Artículo en Inglés | MEDLINE | ID: mdl-39068671

RESUMEN

BACKGROUND: Gain of function disturbances in nutrient sensing are likely the largest component in human age-related disease. Mammalian target of rapamycin complex 1 (mTORC1) activity affects health span and longevity. The drugs ketamine and rapamycin are effective against chronic pain and depression, and both affect mTORC1 activity. Our objective was to measure phosphorylated p70S6K, a marker for mTORC1 activity, in individuals with psychiatric disease to determine whether phosphorylated p70S6K could predict medication response. METHODS: Twenty-seven females provided blood samples in which p70S6K and phosphorylated p70S6K were analyzed. Chart review gathered biometric measurements, clinical phenotypes, and medication response. Questionnaires assessed anxiety, depression, autism traits, and mitochondrial dysfunction, to determine neuropsychiatric disease profiles. Univariate and multivariate statistical analyses were used to identify predictors of medication response. RESULTS: mTORC1 activity correlated highly with both classical biometrics (height, macrocephaly, pupil distance) and specific neuropsychiatric disease profiles (anxiety and autism). Across all cases, phosphorylated p70S6K was the best predictor for ketamine response, and also the best predictor for rapamycin response in a single instance. CONCLUSIONS: The data illustrate the importance of mTORC1 activity in both observable body structure and medication response. This report suggests that a simple assay may allow cost-effective prediction of medication response.


Asunto(s)
Ketamina , Diana Mecanicista del Complejo 1 de la Rapamicina , Proteínas Quinasas S6 Ribosómicas 70-kDa , Humanos , Femenino , Diana Mecanicista del Complejo 1 de la Rapamicina/metabolismo , Persona de Mediana Edad , Ketamina/farmacología , Adulto , Proteínas Quinasas S6 Ribosómicas 70-kDa/metabolismo , Fosforilación , Trastornos Mentales/metabolismo , Sirolimus/farmacología , Sirolimus/uso terapéutico , Monocitos/metabolismo , Monocitos/efectos de los fármacos , Ansiedad/metabolismo , Adulto Joven , Anciano
18.
Toxicol In Vitro ; 100: 105910, 2024 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-39047989

RESUMEN

Per- and Polyfluoroalkyl substances (PFAS) are a group of persistent long-lived chemicals with global environmental contamination. The published literature is rife with confusing and sometimes contradictory effects of PFAS on animal and cell models, as well as epidemiological studies. Cytotoxicity studies are often used as an early indicator to guide safety requirements, regulation, and further studies and thus can be useful to understand important toxicity differences by various PFAS. Recent studies have found that PFAS are not equivalently toxic on all cell types, and that not all cell types exhibit the same sensitivity to individual PFAS. However, immune cells have not been well studied. As immune cells are important for regulating responses to environmental toxins, infection, and cancer, we sought to discover the sensitivity of these cells to various PFAS, including legacy and replacement compounds. We assessed a range of concentrations and found that immune cells are generally more robust when exposed to PFAS, and that Jurkat T-cells were more sensitive than THP-1 monocytes. As monocytes are critical for coordinating inflammatory responses to external threats with cell death cascades, we further investigated these cells. We discovered that THP-1 cells do not undergo organized or programmed death, such as apoptosis or pyroptosis, and instead PFAS exposure results in a more necrotic/lytic and unorganized death, likely contributing to potential inflammatory effects downstream.


Asunto(s)
Supervivencia Celular , Fluorocarburos , Humanos , Fluorocarburos/toxicidad , Supervivencia Celular/efectos de los fármacos , Células Jurkat , Células THP-1 , Contaminantes Ambientales/toxicidad , Monocitos/efectos de los fármacos , Apoptosis/efectos de los fármacos
19.
Thromb Res ; 241: 109092, 2024 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-39024901

RESUMEN

BACKGROUND: Leukocyte-platelet aggregates comprise a pathogenic link between hemostasis and immunity, but the prerequisites and mechanisms of their formation remain not understood. AIMS: To quantify the formation, composition, and morphology of leukocyte-platelet aggregates in vitro under the influence of various cellular activators. METHODS: Phorbol-12-myristate-13-acetate (PMA), lipopolysaccharide (LPS), thrombin receptor-activating peptide (TRAP-6), and adenosine diphosphate (ADP) were used as cellular activators. Flow cytometry was utilized to identify and quantify aggregates in whole human blood and platelet-rich plasma. Cell types and cellular aggregates were identified using fluorescently labeled antibodies against the appropriate cellular markers, and cell activation was assessed by the expression of appropriate surface markers. For confocal fluorescent microscopy, cell membranes and nuclei were labeled. Neutrophil-platelet aggregates were studied using scanning electron microscopy. RESULTS: In the presence of PMA, ADP or TRAP-6, about 17-38 % of neutrophils and 61-77 % of monocytes formed aggregates with platelets in whole blood, whereas LPS did not induce platelet aggregation with either neutrophils or monocytes due the inability to activate platelets. Similar results were obtained when isolated neutrophils were added to platelet-rich plasma. All the cell types involved in the heterotypic aggregation expressed molecular markers of activation. Fluorescent and electron microscopy of the aggregates showed that the predominant platelet/leukocyte ratios were 1:1 and 2:1. CONCLUSIONS: Formation of leukocyte-platelet aggregates depends on the nature of the cellular activator and the spectrum of its cell-activating ability. An indispensable condition for formation of leukocyte-platelet aggregates is activation of all cell types including platelets, which is the restrictive step.


Asunto(s)
Plaquetas , Leucocitos , Lipopolisacáridos , Agregación Plaquetaria , Acetato de Tetradecanoilforbol , Humanos , Plaquetas/metabolismo , Plaquetas/efectos de los fármacos , Acetato de Tetradecanoilforbol/farmacología , Agregación Plaquetaria/efectos de los fármacos , Leucocitos/efectos de los fármacos , Leucocitos/metabolismo , Lipopolisacáridos/farmacología , Adenosina Difosfato/farmacología , Adenosina Difosfato/metabolismo , Neutrófilos/efectos de los fármacos , Neutrófilos/metabolismo , Fragmentos de Péptidos/farmacología , Citometría de Flujo , Monocitos/efectos de los fármacos , Monocitos/metabolismo
20.
Biomed Pharmacother ; 178: 117186, 2024 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-39067165

RESUMEN

In this study, we investigated if the therapeutic potential of peripheral blood mononuclear cell (PBMC) therapy in a murine model of ischemic AKI is related with the survival pattern of monocyte/macrophages in tissue. CD-1 mice were subjected to bilateral renal ischemia followed by reperfusion to induce AKI. M2-polarized PBMCs isolated from CD-1 mice were administered intravenously at different time points post-injury. Our results demonstrate that early administration of PBMC therapy attenuates renal tissue damage, reduces tissue cell death and prevents fibrosis development. Reduction of tissue pyroptosis was observed by reduction on NLRP3 inflammasome activation and decreasing IL-1beta and Caspase-1 expression in the kidney. Furthermore, the therapy was shown to mitigate ferroptosis by inducing GPX4 overexpression. Early administration of PBMCs increased the survival pattern of renal tissue-macrophages, promoting a "pro-survival phenotype" resulting in decreased pyroptotic marker NLRP3, IL-1beta and Caspase 1 and increased anti-ferroptotic gene GPX4. Conversely, delayed administration of PBMC therapy exhibits diminished efficacy in preventing cell death and fibrosis in tissue and provoked a decrease in the pro-survival phenotype of both monocyte /macrophages in tissue. Our findings highlight the therapeutic potential of PBMC therapy in mitigating AKI and preventing CKD progression by modulating tissue-resident macrophage survival and reducing their cell death pathways. The fact that the effectiveness of the therapy depends on the time of administration after the injury underscores the importance of early intervention in AKI management.


Asunto(s)
Lesión Renal Aguda , Fibrosis , Leucocitos Mononucleares , Macrófagos , Monocitos , Animales , Lesión Renal Aguda/patología , Lesión Renal Aguda/metabolismo , Macrófagos/metabolismo , Macrófagos/patología , Macrófagos/efectos de los fármacos , Ratones , Monocitos/metabolismo , Monocitos/efectos de los fármacos , Leucocitos Mononucleares/metabolismo , Masculino , Fosfolípido Hidroperóxido Glutatión Peroxidasa/metabolismo , Ferroptosis/efectos de los fármacos , Piroptosis/efectos de los fármacos , Proteína con Dominio Pirina 3 de la Familia NLR/metabolismo , Riñón/patología , Riñón/efectos de los fármacos , Riñón/metabolismo , Modelos Animales de Enfermedad , Interleucina-1beta/metabolismo , Supervivencia Celular/efectos de los fármacos , Muerte Celular/efectos de los fármacos , Inflamasomas/metabolismo , Caspasa 1/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA