Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Resultados 1 - 20 de 7.435
Filtrar
Más filtros

Publication year range
1.
Blood ; 141(6): 659-672, 2023 02 09.
Artículo en Inglés | MEDLINE | ID: mdl-36201744

RESUMEN

Relapse limits the therapeutic efficacy both of chimeric antigen receptor (CAR) T cells and allogeneic hematopoietic cell transplantation (allo-HCT). Patients may undergo these therapies sequentially to prevent or treat relapsed malignancy. However, direct integration of the 2 therapies has been avoided over concerns for potential induction of graft-versus-host disease (GVHD) by allogeneic CAR T cells. We have shown in murine T-cell-replete MHC-haploidentical allo-HCT that suppressive mechanisms induced immediately after posttransplant cyclophosphamide (PTCy), given on days +3/+4, prevent GVHD induction by alloreactive T cells infused as early as day +5. Therefore, we hypothesized that allogeneic CAR T cells given in a similarly integrated manner in our murine MHC-haploidentical allo-HCT model may safely exert antitumor effects. Indeed, allogeneic anti-CD19 CAR T cells given early after (day +5) PTCy or even prior to (day 0) PTCy cleared leukemia without exacerbating the cytokine release syndrome occurring from the MHC-haploidentical allo-HCT or interfering with PTCy-mediated GVHD prevention. Meanwhile, CAR T-cell treatment on day +9 or day +14 was safe but less effective, suggesting a limited therapeutic window. CAR T cells infused before PTCy were not eliminated, but surviving CAR T cells continued to proliferate highly and expand despite PTCy. In comparison with infusion on day +5, CAR T-cell infusion on day 0 demonstrated superior clinical efficacy associated with earlier CAR T-cell expansion, higher phenotypic CAR T-cell activation, less CD4+CD25+Foxp3+ CAR T-cell recovery, and transcriptional changes suggesting increased activation of CD4+ CAR T cells and more cytotoxic CD8+ CAR T cells. This study provides mechanistic insight into PTCy's impact on graft-versus-tumor immunity and describes novel approaches to integrate CAR T cells and allo-HCT that may compensate for deficiencies of each individual approach.


Asunto(s)
Enfermedad Injerto contra Huésped , Trasplante de Células Madre Hematopoyéticas , Leucemia , Humanos , Ratones , Animales , Ciclofosfamida/farmacología , Ciclofosfamida/uso terapéutico , Trasplante de Células Madre Hematopoyéticas/efectos adversos , Linfocitos T CD4-Positivos/patología , Leucemia/tratamiento farmacológico
2.
Immunity ; 45(4): 714-716, 2016 10 18.
Artículo en Inglés | MEDLINE | ID: mdl-27760335

RESUMEN

It has recently become apparent that the gut microbiota modulates the response to cancer therapy. In this issue of Immunity, Daillère et al. (2016) identified two bacterial species potentiating the anti-tumor effect of cyclophosphamide that are kept in check by the sensor NOD2.


Asunto(s)
Bacterias/metabolismo , Microbioma Gastrointestinal/fisiología , Neoplasias/microbiología , Proteína Adaptadora de Señalización NOD2/metabolismo , Animales , Ciclofosfamida/farmacología , Ciclofosfamida/uso terapéutico , Humanos , Neoplasias/tratamiento farmacológico
3.
Immunity ; 45(4): 931-943, 2016 10 18.
Artículo en Inglés | MEDLINE | ID: mdl-27717798

RESUMEN

The efficacy of the anti-cancer immunomodulatory agent cyclophosphamide (CTX) relies on intestinal bacteria. How and which relevant bacterial species are involved in tumor immunosurveillance, and their mechanism of action are unclear. Here, we identified two bacterial species, Enterococcus hirae and Barnesiella intestinihominis that are involved during CTX therapy. Whereas E. hirae translocated from the small intestine to secondary lymphoid organs and increased the intratumoral CD8/Treg ratio, B. intestinihominis accumulated in the colon and promoted the infiltration of IFN-γ-producing γδT cells in cancer lesions. The immune sensor, NOD2, limited CTX-induced cancer immunosurveillance and the bioactivity of these microbes. Finally, E. hirae and B. intestinihominis specific-memory Th1 cell immune responses selectively predicted longer progression-free survival in advanced lung and ovarian cancer patients treated with chemo-immunotherapy. Altogether, E. hirae and B. intestinihominis represent valuable "oncomicrobiotics" ameliorating the efficacy of the most common alkylating immunomodulatory compound.


Asunto(s)
Ciclofosfamida/farmacología , Enterococcus hirae/inmunología , Factores Inmunológicos/inmunología , Neoplasias/tratamiento farmacológico , Neoplasias/inmunología , Animales , Colon/inmunología , Colon/microbiología , Memoria Inmunológica/inmunología , Inmunoterapia/métodos , Interferón gamma/inmunología , Intestino Delgado/inmunología , Intestino Delgado/microbiología , Ratones , Ratones Endogámicos C57BL , Monitorización Inmunológica , Proteína Adaptadora de Señalización NOD2/inmunología , Células TH1/inmunología
4.
Antimicrob Agents Chemother ; 68(8): e0152023, 2024 Aug 07.
Artículo en Inglés | MEDLINE | ID: mdl-38990014

RESUMEN

Mycobacterium abscessus pulmonary infections are increasingly problematic, especially for immunocompromised individuals and those with underlying lung conditions. Currently, there is no reliable standardized treatment, underscoring the need for improved preclinical drug testing. We present a simplified immunosuppressed mouse model using only four injections of cyclophosphamide, which allows for sustained M. abscessus lung burden for up to 16 days. This model proved effective for antibiotic efficacy evaluation, as demonstrated with imipenem or amikacin.


Asunto(s)
Amicacina , Antibacterianos , Ciclofosfamida , Modelos Animales de Enfermedad , Infecciones por Mycobacterium no Tuberculosas , Mycobacterium abscessus , Animales , Ciclofosfamida/farmacología , Mycobacterium abscessus/efectos de los fármacos , Ratones , Infecciones por Mycobacterium no Tuberculosas/tratamiento farmacológico , Infecciones por Mycobacterium no Tuberculosas/microbiología , Antibacterianos/farmacología , Antibacterianos/uso terapéutico , Amicacina/farmacología , Amicacina/uso terapéutico , Imipenem/farmacología , Imipenem/uso terapéutico , Pulmón/microbiología , Pulmón/efectos de los fármacos , Huésped Inmunocomprometido , Femenino
5.
Biochem Biophys Res Commun ; 734: 150777, 2024 11 19.
Artículo en Inglés | MEDLINE | ID: mdl-39383831

RESUMEN

Syringic acid (SA), a naturally occurring phenolic substance present in many edible plants and fruits, has been shown to have potential in immunoenhancement applications. In this study, we investigated the immunomodulatory effects of SA in mitigating cyclophosphamide (CYP)-induced immunosuppression in BALB/c mice using doxycycline as a positive control. SA administration prevented immune organ atrophy and morphological changes in the thymus, spleen, and bone marrow induced by CYP treatment in mice while also showing a dose-dependent enhancement of thymus and spleen indices compared to mice treated with CYP alone. Furthermore, SA improved thymocyte and splenocyte proliferation and exhibited significant antioxidant activity by reducing the elevated levels of malondialdehyde induced by CYP treatment. SA treatment effectively restored white blood cell (WBC) and lymphocyte counts to normal levels in CYP-treated animals, and the protective effects of CYP on immunological tissues were confirmed through histopathological examination. Moreover, SA treatment upregulated the expression of IL-6, IL-7, IL-15, and FoxN1. Finally, molecular docking studies revealed that binding energy values predicted minor inhibition potential toward IL-6, IL-7, FoxN1, IL-15, STAT3, STAT5, and JAK3. Overall, our findings suggest that SA treatment has the potential to reduce CYP-induced immunosuppression and may have applications as an immunologic adjuvant or functional food additive in chemotherapy.


Asunto(s)
Ciclofosfamida , Ácido Gálico , Ratones Endogámicos BALB C , Animales , Ciclofosfamida/farmacología , Ácido Gálico/análogos & derivados , Ácido Gálico/farmacología , Ácido Gálico/química , Ratones , Timo/efectos de los fármacos , Timo/patología , Timo/inmunología , Timo/metabolismo , Terapia de Inmunosupresión , Bazo/efectos de los fármacos , Bazo/metabolismo , Bazo/inmunología , Inmunosupresores/farmacología , Simulación del Acoplamiento Molecular , Citocinas/metabolismo , Masculino , Antioxidantes/farmacología , Modelos Animales de Enfermedad , Proliferación Celular/efectos de los fármacos , Factores de Transcripción Forkhead/metabolismo
6.
J Comput Chem ; 45(10): 663-670, 2024 Apr 15.
Artículo en Inglés | MEDLINE | ID: mdl-38088485

RESUMEN

The oxazaphosphorine cyclophosphamide (CP) is a DNA-alkylating agent commonly used in cancer chemotherapy. This anticancer agent is administered as a prodrug activated by a liver cytochrome P450-catalyzed 4-hydroxylation reaction that yields the active, cytotoxic metabolite. The primary metabolite, 4-hydroxycyclophosphamide, equilibrates with the ring-open aldophosphamide that undergoes ß-elimination to yield the therapeutically active DNA cross-linking phosphoramide mustard and the byproduct acrolein. The present paper presents a DFT investigation of the different metabolic phases and an insight into the mechanism by which CP exerts its cytotoxic action. A detailed computational analysis of the energy profiles describing all the involved transformations and the mechanism of DNA alkylation is given with the aim to contribute to an increase of knowledge that, after more than 60 years of unsuccessful attempts, can lead to the design and development of a new generation of oxazaphosphorines.


Asunto(s)
Acroleína , ADN , Ciclofosfamida/farmacología , Hidroxilación
7.
Reprod Biol Endocrinol ; 22(1): 125, 2024 Oct 16.
Artículo en Inglés | MEDLINE | ID: mdl-39415205

RESUMEN

BACKGROUND: Restoring the function of the ovary is important for chemotherapy-induced ovarian failure (COF) patients. Stem cell and extracellular vesicles (EVs) therapy show promise but need further improvement. METHODS: Human umbilical cord mesenchymal stem cells (hUC-MSCs) were primarily cultured and further three-dimensional (3D) cultured using an ultra-low attachment surface method. The expression levels of nutritional cytokines and immunomodulatory and stemness-related genes of 3D-cultured hUC-MSCs were analyzed. EVs were isolated by ultracentrifugation and characterized. Ovaries were decellularized with sodium dodecyl sulfate to obtain extracellular matrix (ECM). Lyophilized EVs from three-dimensional (2D) or 3D hUC-MSCs were mixed with ECM to prepare the 2D/3D-MSC-EVs-ECM gels. The therapeutic effect of the MSC-EVs-ECM gel on cyclophosphamide (CTX) -treated rats was analyzed through various tests. RNA sequencing was used to analyze the expression changes of genes before and after treatment. RESULTS: After culturing in ultra-low attachment dishes, hUC-MSCs aggregated into spheroids and significantly upregulated the expression levels of immunomodulatory and stemness-related genes. The total EVs yield was also upregulated (5.6-fold) after 3D culture. The cell viability of CTX-treated ovarian granulosa cells (OGCs) was significantly rescued by coculture with the 3D-MSC-EVs-ECM gel. Hormones indicative of ovarian function, AMH, E2, and FSH, were recovered in both the CTX + 2D-MSC-EVs-ECM gel group and the CTX + 3D-MSC-EVs-ECM gel group, while the apoptosis-related protein Bax was significantly downregulated. The 3D-MSC-EVs-ECM gel was more effective than the 2D-MSC-EVs-ECM gel. Significantly differentially expressed genes, such as Hbb-b1, Gpd1, and Sirpa, were detected by RNA sequencing. Hbb-b1 was increased in the ovaries of CTX-treated rats, and this increase was attenuated by injecting the 2D/3D-MSC-EVs-ECM gel. Gpd1 was increased after CTX treatment, and this increase was reversed by the 3D-MSC-EVs-ECM gel. Sirpa was decreased in the ovaries of CTX-treated rats, and this decrease was attenuated by injecting the 3D-MSC-EVs-ECM gel. CONCLUSIONS: Our study demonstrated that the 3D-MSC-EVs-ECM gel is an efficient strategy for the recovery of ovarian function in CTX-induced ovarian failure.


Asunto(s)
Vesículas Extracelulares , Células Madre Mesenquimatosas , Ovario , Femenino , Animales , Ratas , Células Madre Mesenquimatosas/metabolismo , Células Madre Mesenquimatosas/efectos de los fármacos , Humanos , Ovario/efectos de los fármacos , Ovario/metabolismo , Vesículas Extracelulares/metabolismo , Vesículas Extracelulares/efectos de los fármacos , Matriz Extracelular/metabolismo , Matriz Extracelular/efectos de los fármacos , Trasplante de Células Madre Mesenquimatosas/métodos , Ratas Sprague-Dawley , Ciclofosfamida/farmacología , Geles , Células Cultivadas , Insuficiencia Ovárica Primaria/terapia , Técnicas de Cultivo Tridimensional de Células/métodos
8.
Chem Senses ; 492024 01 01.
Artículo en Inglés | MEDLINE | ID: mdl-38421250

RESUMEN

Many common chemotherapeutics produce disruptions in the sense of taste which can lead to loss of appetite, nutritional imbalance, and reduced quality of life, especially if taste loss persists after treatment ends. Cyclophosphamide (CYP), an alkylating chemotherapeutic agent, affects taste sensitivity through its cytotoxic effects on mature taste receptor cells (TRCs) and on taste progenitor cell populations, retarding the capacity to replace TRCs. Mechanistic studies have focused primarily on taste cells, however, taste signaling requires communication between TRCs and the gustatory nerve fibers that innervate them. Here, we evaluate cyclophosphamide's effects on the peripheral gustatory nerve fibers that innervate the taste buds. Following histological analysis of tongue tissues, we find that CYP reduces innervation within the fungiform and circumvallates taste buds within 4 days after administration. To better understand the dynamics of the denervation process, we used 2-photon intravital imaging to visualize the peripheral gustatory nerve fibers within individual fungiform taste buds up to 20 days after CYP treatment. We find that gustatory fibers retract from the taste bud properly but are maintained within the central papilla core. These data indicate that in addition to TRCs, gustatory nerve fibers are also affected by CYP treatment. Because the connectivity between TRCs and gustatory neurons must be re-established for proper function, gustatory fibers should continue to be included in future studies to understand the mechanisms leading to chemotherapy-induced persistent taste loss.


Asunto(s)
Ageusia , Papilas Gustativas , Animales , Ratones , Papilas Gustativas/fisiología , Calidad de Vida , Lengua , Ciclofosfamida/farmacología , Gusto
9.
Chem Res Toxicol ; 37(2): 259-273, 2024 02 19.
Artículo en Inglés | MEDLINE | ID: mdl-38183658

RESUMEN

Resorcinolic lipids are described as potential examples of selective chemotherapeutic adjuvants that can enhance the effects of cyclophosphamide (CYC) while promoting cell death without causing DNA damage. Therefore, the current study attempted to describe how the resorcinolic lipid methyl 3,5-dimethoxy-2-octanoylbenzoate (AMS35BB) interacted with DNA (DNA docking) and how this compound affected genetic toxicology models and other biological characteristics when combined with CYC. We observed that AMS35BB, used alone (7.5 and 10 mg/kg), increases the frequency of genomic damage (comet assay) but not chromosomal damage (micronuclei assay), lowers phagocytosis, and promotes cell death in Swiss male mice. When used in association with CYC, AMS35BB can reduce the risk of genomic damage by up to 33.8% as well as chromosomal damage, splenic phagocytosis, cell death, and lymphocyte frequency. Molecular docking showed that AMS35BB had a higher affinity than the active metabolite of CYC for binding to the DNA double helix major groove. As a result, AMS35BB has the potential to be both an adjuvant when used in association with CYC and a therapeutic candidate for the development of a selective chemotherapeutic drug.


Asunto(s)
ADN , Ratones , Animales , Masculino , Simulación del Acoplamiento Molecular , Ciclofosfamida/farmacología , Muerte Celular , Ensayo Cometa
10.
Reprod Biomed Online ; 49(3): 103914, 2024 09.
Artículo en Inglés | MEDLINE | ID: mdl-38917774

RESUMEN

RESEARCH QUESTION: What impact does dehydroepiandrosterone (DHEA) have on ovarian angiogenesis and function in a rat model of with premature ovarian insufficiency (POI), and what are the potential mechanisms of action? DESIGN: DHEA was added to a culture of human microvascular endothelial cells (HMEC-1) to investigate its effects on cell proliferation, migration and tube formation. A rat model of POI was established by intraperitoneal injection of cyclophosphamide, followed by continuous oral administration of DHEA or vehicle for 28 days. Ovarian angiogenesis, follicular growth and granulosa cell survival in ovarian tissues were assessed through haematoxylin and eosin staining, immunohistochemistry and TdT (terminal deoxynucleotidyl transferase)-mediated dUTP nick-end labelling (TUNEL). The effect of DHEA on the fertility of rats with POI was evaluated in pregnant animals. The expression levels of characteristic genes and proteins in the hypoxia-inducible factor (HIF)-1α/vascular endothelial growth factor (VEGF) pathway was determined using quantitative reverse transcription PCR and western blotting. RESULTS: In-vitro experiments revealed that DHEA stimulated the proliferation, migration and tube formation of HMEC-1. In in-vivo studies, DHEA treatment improved the disruption of the oestrous cycle and hormone imbalances in POI rats. Key genes in the HIF-1α/VEGF pathway exhibited up-regulated expression, promoting ovarian angiogenesis in POI rats, and enhancing follicular development and granulosa cell survival, thereby restoring fertility in rats. CONCLUSIONS: DHEA can potentially restore ovarian function in rats with cyclophosphamide-induced POI by up-regulating HIF-1α/VEGF signalling, which promotes the growth of blood vessels in the ovaries.


Asunto(s)
Deshidroepiandrosterona , Modelos Animales de Enfermedad , Subunidad alfa del Factor 1 Inducible por Hipoxia , Ovario , Insuficiencia Ovárica Primaria , Transducción de Señal , Factor A de Crecimiento Endotelial Vascular , Animales , Femenino , Insuficiencia Ovárica Primaria/tratamiento farmacológico , Deshidroepiandrosterona/farmacología , Subunidad alfa del Factor 1 Inducible por Hipoxia/metabolismo , Ovario/irrigación sanguínea , Ovario/efectos de los fármacos , Ratas , Factor A de Crecimiento Endotelial Vascular/metabolismo , Transducción de Señal/efectos de los fármacos , Neovascularización Fisiológica/efectos de los fármacos , Ratas Sprague-Dawley , Humanos , Regulación hacia Arriba/efectos de los fármacos , Proliferación Celular/efectos de los fármacos , Movimiento Celular/efectos de los fármacos , Embarazo , Ciclofosfamida/farmacología , Angiogénesis
11.
Mol Cell Probes ; 76: 101969, 2024 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-38964425

RESUMEN

The progression and pathogenesis of membranous glomerulonephritis (MGN) are inextricably linked to chronic inflammation. Despite improving clinical remission rates due to the application of cyclophosphamide (CYC), treatment of MGN still requires further exploration. Ruxolitinib (Ruxo) negatively affects the signaling pathways participating in the production of pro-inflammatory cytokines. Hence, we investigated whether the combination of CYC and Ruxo can modulate inflammation through influencing T helper 17 (Th17) lineages and regulatory T cells (Tregs). Passive Heymann nephritis (PHN), an experimental model of MGN, was induced in a population of rats. Then, the animals were divided into five groups: PHN, CYC-receiving, Ruxo-receiving, CYC-Ruxo-receiving PHN rats, and healthy controls. After 28 days of treatment, biochemistry analysis was performed and splenocytes were isolated for flowcytometry investigation of Th17 cells and Tregs. The correlative transcription factors of the cells, alongside their downstream cytokine gene expressions, were also assessed using real-time PCR. Furthermore, serum cytokine signatures for the lymphocytes were determined through ELISA. The combination of CYC and Ruxo significantly reduced the serum values of urea in rats versus the PHN group (24.62 ± 7.970 vs. 40.60 ± 10.81 mg/dL). In contrast to Treg's activities, the functionality of Th17 cells noticeably increased not only in PHN rats but also in CYC or Ruxo-receiving PHN animals when compared with the control (10.60 ± 2.236, 8.800 ± 1.465, 8.680 ± 1.314 vs. 4.420 ± 1.551 %). However, in comparison to the PHN group, the incidence of Th17 cells notably fell in rats receiving CYC and Ruxo (10.60 ± 2.236 vs. 6.000 ± 1.373 %) in favor of the Treg's percentage (5.020 ± 1.761 vs. 8.980 ± 1.178 %), which was verified by the gene expressions and cytokine productions correlative to these lymphocytes. The combination of CYC and Ruxo was able to decline Th17 cells in favor of Tregs improvement in PHN rats, suggesting an innovative combination therapy in MGN treatment approaches.


Asunto(s)
Ciclofosfamida , Citocinas , Glomerulonefritis Membranosa , Nitrilos , Pirazoles , Pirimidinas , Linfocitos T Reguladores , Células Th17 , Animales , Glomerulonefritis Membranosa/tratamiento farmacológico , Glomerulonefritis Membranosa/inmunología , Linfocitos T Reguladores/efectos de los fármacos , Linfocitos T Reguladores/inmunología , Células Th17/efectos de los fármacos , Células Th17/inmunología , Ciclofosfamida/farmacología , Ciclofosfamida/uso terapéutico , Nitrilos/farmacología , Nitrilos/uso terapéutico , Pirimidinas/farmacología , Pirimidinas/uso terapéutico , Ratas , Pirazoles/farmacología , Pirazoles/uso terapéutico , Citocinas/metabolismo , Masculino , Modelos Animales de Enfermedad , Quimioterapia Combinada
12.
Mol Biol Rep ; 51(1): 930, 2024 Aug 22.
Artículo en Inglés | MEDLINE | ID: mdl-39174728

RESUMEN

BACKGROUND: Among the three most used anticancer drugs, cyclophosphamide, Adriamycin, and 5-Fluorouracil (CAF), the most significant outcome is chemobrain, caused by increased oxidative stress, inflammatory insult, and mitochondrial dysfunction. OBJECTIVE: In this study, endogenous antioxidant coenzyme Q10 (CoQ10) was evaluated for its neuroprotective effects in CICI. MATERIALS AND METHODS: The chemobrain was induced in Swiss albino female mice by administering CAF (40 + 4 + 25 mg/kg) intraperitoneal (i.p.) in three cycles (single injection per week) followed by treatment with CoQ10 (40 mg/kg; p.o.) for up to 3 weeks followed by behavioral, biochemical, molecular and histopathological analysis. RESULTS: Treatment with CoQ10 significantly improved cognition by improving exploring time in novel objects recognition test followed by increasing the time spent in the target quadrant in MWM test as compared to CAF-treated animals. Moreover, CoQ10 demonstrated antioxidant properties by reducing the expression of LPO while increasing levels of GSH, SOD, and catalase as compared to CAF-treated animals. While the levels of AChEs were significantly reduced after CoQ10 treatment in CAF-treated animals. In terms of its mechanism, it effectively counteracted the pro-inflammatory substances (TNF-α and IL-1ß) triggered by CAF while also enhancing the levels of anti-inflammatory markers (IL-10 and Nrf2). Moreover, CoQ10 showed mitochondrial enhancers and it improved the level of Complex (I, II, and IV). Besides that, mitochondrial morphological analysis was done by TEM, and neuronal morphology along with quantification analysis was performed by H&E staining using Image J software to confirm the neuroprotective effect of CoQ10 over CAF-induced cognitive impairment. CONCLUSION: This study suggests CoQ10 can protect the mitochondria by imposing antioxidant, and anti-inflammatory properties, which could be a potential therapy for CICI.


Asunto(s)
Antioxidantes , Estrés Oxidativo , Ubiquinona , Animales , Ubiquinona/análogos & derivados , Ubiquinona/farmacología , Ubiquinona/uso terapéutico , Ratones , Femenino , Antioxidantes/farmacología , Estrés Oxidativo/efectos de los fármacos , Deterioro Cognitivo Relacionado con la Quimioterapia/tratamiento farmacológico , Deterioro Cognitivo Relacionado con la Quimioterapia/metabolismo , Disfunción Cognitiva/tratamiento farmacológico , Disfunción Cognitiva/inducido químicamente , Disfunción Cognitiva/metabolismo , Fármacos Neuroprotectores/farmacología , Fármacos Neuroprotectores/uso terapéutico , Doxorrubicina/efectos adversos , Fluorouracilo/efectos adversos , Fluorouracilo/farmacología , Modelos Animales de Enfermedad , Antineoplásicos/farmacología , Antineoplásicos/efectos adversos , Ciclofosfamida/efectos adversos , Ciclofosfamida/farmacología
13.
Bioorg Chem ; 151: 107709, 2024 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-39137599

RESUMEN

Ginger is an important cooking spice and herb worldwide, and scientific research has gradually confirmed the effect of ginger on preventing hair loss. Cedrol (CE) is a small sesquiterpene molecule in ginger and its external administration (EA) has shown hope in promoting hair growth, and alternative administration mode has become a potential treatment scheme to improve the efficacy of CE. The purpose of this study is to evaluate the effects of oral administration (OA) and EA of CE on hair regeneration of C57BL/6 alopecia areata (AA) mice induced by cyclophosphamide (CP) and to clarify the potential hair growth mechanism of CE in AA model in vitro and in vivo. The results showed that CE-OA has a shorter hair-turning black time and faster hair growth rate, and can lessen hair follicle damage induced by CP and promote hair follicle cell proliferation. Its effect is superior to CE-EA. At the same time, CE can increase the cytokines IFN-γ, IL-2, and IL-7 in the serum of mice, and decrease the expression of adhesion factors ICAM-1 and ELAM-1, thus alleviating the immunosuppression induced by CP. Mechanism research shows that CE regulates the JAK3/STAT3 signaling pathway, activates the Wnt3α/ß-catenin germinal center, and ameliorates oxidative stress induced by CP, thus promoting the proliferation of hair follicle cells and reversing AA. These results provide a theoretical basis for understanding the anti-AA mechanism of CE-OA, indicating that CE can be used as raw material for developing oral hair growth drugs.


Asunto(s)
Ratones Endogámicos C57BL , Sesquiterpenos , Zingiber officinale , Animales , Zingiber officinale/química , Administración Oral , Ratones , Sesquiterpenos/farmacología , Sesquiterpenos/química , Cabello/efectos de los fármacos , Cabello/química , Proliferación Celular/efectos de los fármacos , Regeneración/efectos de los fármacos , Folículo Piloso/efectos de los fármacos , Folículo Piloso/metabolismo , Estructura Molecular , Masculino , Relación Dosis-Respuesta a Droga , Alopecia Areata/tratamiento farmacológico , Relación Estructura-Actividad , Ciclofosfamida/farmacología , Sesquiterpenos Policíclicos
14.
Mar Drugs ; 22(6)2024 May 21.
Artículo en Inglés | MEDLINE | ID: mdl-38921545

RESUMEN

Deep seawater (DS), obtained from a depth over 200 m, has health benefits due to its rich nutrients and minerals, and intake of DS has shown diverse immunomodulatory effects in allergies and cancer. Therefore, the immunostimulatory effects of Korean mineral-rich seawaters were examined in a cyclophosphamide (CPA)-induced immunosuppression model. Three samples of Korean seawater, namely DS from the East Sea off the coasts of Pohang (PDS) and Uljin (UDS), and seawater from the West Sea off the coast of Boryeong (BS), were collected. The seawaters were abundant in several minerals (calcium, iron, zinc, selenium, etc.). Mice were orally administered the seawaters for 42 days, followed by CPA-induced immunosuppression. The CPA induction reduced the weight of the spleen and lymph nodes; however, the administration of seawaters increased the weight of the lymphoid organs, accompanied by stimulation of natural killer cells' activity and NF-kB-mediated cytokine production (IFNγ, TNFα, IL1ß, IL6, and IL12). The mouse-derived splenocytes showed lymphoproliferation without cytotoxicity in the seawater groups. Histopathological analysis revealed that the seawaters improved the CPA-induced atrophic changes by promoting lymphoproliferation in the spleen and lymph nodes. These results provide useful information for the use of Korean mineral-rich seawaters, particularly PDS and UDS, as alternative immunostimulants under immunosuppressive conditions.


Asunto(s)
Ciclofosfamida , Agua de Mar , Animales , Ciclofosfamida/farmacología , Ratones , Minerales/farmacología , Citocinas/metabolismo , República de Corea , Terapia de Inmunosupresión , Bazo/efectos de los fármacos , Bazo/inmunología , Células Asesinas Naturales/efectos de los fármacos , Células Asesinas Naturales/inmunología , Masculino , Adyuvantes Inmunológicos/farmacología , Ganglios Linfáticos/efectos de los fármacos , Ganglios Linfáticos/inmunología , Inmunosupresores/farmacología , Ratones Endogámicos BALB C
15.
Environ Toxicol ; 39(3): 1315-1322, 2024 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-37955270

RESUMEN

In this study, it was aimed to investigate the effects of berberine (BER) and curcumin (CUR) in the experimental model of cystitis induced by cyclophosphamide (CYP). A total of 36 Wistar-Albino female rats were used in the study. Rats were randomly divided into six groups (n = 6). Normal control group, dimethyl sulfoxide (DMSO) group, CYP group (75 mg/kg), CYP + BER (75 mg/kg CYP and 50 mg/kg BER), CYP + CUR group (75 mg/kg CYP and 50 mg/kg CUR), CYP + BER + CUR group (75 mg/kg CYP and 50 mg/kg BER and 50 mg/kg CUR). Severe edema, hyperemia, hemorrhage, necrosis, and thinning of the epithelial layer were observed in the CYP group. BER and CUR treatment significantly reduced these pathologies. Masson-Trichrome staining was severe in the CYP group and moderate in the CYP + BER, CYP + CUR, and CYP + BER + CUR groups. In the CYP group, there was a severe expression of caspase-3, TNF-α and IL-6, and mild expression of IL-10. BER and CUR treatment decreased the expression of caspase-3, TNF-α, and IL-6 and increased the expression of IL-10. The findings of the study reveal that BER and CUR treatments may reduce CYP-induced bladder damage by reducing apoptosis and inflammation and ameliorating histopathological changes.


Asunto(s)
Berberina , Curcumina , Cistitis Intersticial , Ratas , Animales , Curcumina/farmacología , Interleucina-10 , Caspasa 3 , Ratas Wistar , Factor de Necrosis Tumoral alfa , Interleucina-6 , Ciclofosfamida/farmacología
16.
Int J Mol Sci ; 25(16)2024 Aug 13.
Artículo en Inglés | MEDLINE | ID: mdl-39201507

RESUMEN

Metronomic chemotherapy with cyclophosphamide (Cpp) has shown promising results in cancer protocols. These lower and prolonged doses have antiangiogenic, pro-cytotoxic, and moderate secondary effects. Molecular iodine (I2) reduces the viability of cancer cells and, with chemotherapeutic agents, activates the antitumoral immune response and diminishes side effects. The present work evaluates the adjuvant of oral I2 with Cpp using a murine model of mammary cancer. Female Sprague Dawley rats with 7,12-dimethylbenzantracene-induced tumors received Cpp intraperitoneal (50 and 70 mg/kg two times/week, iCpp50 and iCpp70) and oral (0.03%; 50 mg/Kg; oCpp50) doses. I2 (0.05%, 50 mg/100 mL) and oCpp50 were offered in drinking water for three weeks. iCpp70 was the most efficient antitumoral dose but generated severe body weight loss and hemorrhagic cystitis (HC). I2 prevented body weight loss, exhibited adjuvant actions with Cpp, decreasing tumor growth, and canceled HC mechanisms, including decreases in vascular endothelial growth factor (VEGF) and Survivin expression. oCpp50 + I2 diminished angiogenic signals (CD34, vessel-length, and VEGF content) and proinflammatory cytokines (interleukin-10 and tumor necrosis factor-alpha) and increased cytotoxic (lymphocytic infiltration, CD8+ cells, Tbet, and interferon-gamma) and antioxidant markers (nuclear erythroid factor-2 and glutathione peroxidase). I2 enhances the effectiveness of oCpp, making it a compelling candidate for a clinical protocol.


Asunto(s)
Administración Metronómica , Ciclofosfamida , Yodo , Ratas Sprague-Dawley , Animales , Ciclofosfamida/farmacología , Ciclofosfamida/efectos adversos , Ciclofosfamida/administración & dosificación , Femenino , Ratas , Yodo/administración & dosificación , Yodo/farmacología , Neoplasias Mamarias Experimentales/tratamiento farmacológico , Neoplasias Mamarias Experimentales/patología , Neoplasias Mamarias Experimentales/metabolismo , Factor A de Crecimiento Endotelial Vascular/metabolismo , Factor A de Crecimiento Endotelial Vascular/genética , Progresión de la Enfermedad , Antineoplásicos Alquilantes/efectos adversos , Antineoplásicos Alquilantes/administración & dosificación , Antineoplásicos Alquilantes/farmacología
17.
Int J Mol Sci ; 25(14)2024 Jul 20.
Artículo en Inglés | MEDLINE | ID: mdl-39063180

RESUMEN

T-cell malignancies, including T-cell acute lymphoblastic leukemia (T-ALL) and T-cell lymphoblastic lymphoma (T-LBL), present significant challenges to treatment due to their aggressive nature and chemoresistance. Chemotherapies remain a mainstay for their management, but the aggressiveness of these cancers and their associated toxicities pose limitations. Immunepotent CRP (ICRP), a bovine dialyzable leukocyte extract, has shown promise in inducing cytotoxicity against various cancer types, including hematological cancers. In this study, we investigated the combined effect of ICRP with a panel of chemotherapies on cell line models of T-ALL and T-LBL (CEM and L5178Y-R cells, respectively) and its impact on immune system cells (peripheral blood mononuclear cells, splenic and bone marrow cells). Our findings demonstrate that combining ICRP with chemotherapies enhances cytotoxicity against tumoral T-cell lymphoblasts. ICRP + Cyclophosphamide (CTX) cytotoxicity is induced through a caspase-, reactive oxygen species (ROS)-, and calcium-dependent mechanism involving the loss of mitochondrial membrane potential, an increase in ROS production, and caspase activation. Low doses of ICRP in combination with CTX spare non-tumoral immune cells, overcome the bone marrow-induced resistance to CTX cell death, and improves the CTX antitumor effect in vivo in syngeneic Balb/c mice challenged with L5178Y-R. This led to a reduction in tumor volume and a decrease in Ki-67 proliferation marker expression and the granulocyte/lymphocyte ratio. These results set the basis for further research into the clinical application of ICRP in combination with chemotherapeutic regimens for improving outcomes in T-cell malignancies.


Asunto(s)
Ciclofosfamida , Especies Reactivas de Oxígeno , Animales , Ratones , Humanos , Línea Celular Tumoral , Especies Reactivas de Oxígeno/metabolismo , Ciclofosfamida/farmacología , Sinergismo Farmacológico , Potencial de la Membrana Mitocondrial/efectos de los fármacos , Leucemia-Linfoma Linfoblástico de Células T Precursoras/tratamiento farmacológico , Leucemia-Linfoma Linfoblástico de Células T Precursoras/metabolismo , Leucemia-Linfoma Linfoblástico de Células T Precursoras/patología , Apoptosis/efectos de los fármacos , Bovinos , Muerte Celular/efectos de los fármacos , Antineoplásicos/farmacología , Linfocitos T/efectos de los fármacos , Linfocitos T/inmunología , Linfocitos T/metabolismo , Ratones Endogámicos BALB C , Proliferación Celular/efectos de los fármacos
18.
Bull Exp Biol Med ; 177(3): 318-322, 2024 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-39138791

RESUMEN

We studied the expression of Nrf2 transcription factor and antioxidant system proteins in drug-resistant murine leukemia strains P388 in vivo, as well as the redox status of cells under conditions of induced oxidative stress. Immunoblotting and real-time PCR showed that the cyclophosphamide-resistant strain P388 (P388/CP) exhibits Nrf2-mediated drug resistance. Cells of the P388/CP strain are characterized by high expression of Nrf2, which leads to a significant increase in the expression of ARE genes and antioxidant system proteins, as well as to the effective maintenance of redox homeostasis under conditions of induced oxidative stress. Taking into account the important role of Nrf2 overexpression in reducing the effectiveness of chemotherapy in patients with different leukemias, the P388/CP strain can be of great interest as a model in the development of new drugs for the treatment of malignant neoplasms.


Asunto(s)
Antioxidantes , Resistencia a Antineoplásicos , Factor 2 Relacionado con NF-E2 , Estrés Oxidativo , Animales , Factor 2 Relacionado con NF-E2/metabolismo , Factor 2 Relacionado con NF-E2/genética , Ratones , Resistencia a Antineoplásicos/genética , Resistencia a Antineoplásicos/efectos de los fármacos , Estrés Oxidativo/efectos de los fármacos , Antioxidantes/farmacología , Leucemia P388/tratamiento farmacológico , Leucemia P388/metabolismo , Leucemia P388/genética , Leucemia P388/patología , Ciclofosfamida/farmacología , Oxidación-Reducción/efectos de los fármacos
19.
J Cell Mol Med ; 27(13): 1880-1886, 2023 07.
Artículo en Inglés | MEDLINE | ID: mdl-37246626

RESUMEN

Dealing with nude mice, which lack thymus and therefore are sensitive to unsterile conditions, needs special care and laboratory conditions. For preclinical studies, especially tumour imaging purposes, in which therapeutic properties of drugs or therapeutic compounds are not studied, mice with normal immune system can be a favourable alternative if they carry tumours of interest. In the current study, we introduce an optimized protocol for induction of human tumours in BALB/c mice for preclinical studies. Immune system of BALB/c mice was suppressed by administration of cyclosporine A (CsA), ketoconazole and cyclophosphamide. The tumours of MDA-MB-231, A-431 and U-87-MG human cancer cells were induced by subcutaneous injection of the cells to the immunosuppressed mice. Tumour size was calculated weekly. Histopathological and metastatic analyses were performed using haematoxylin and eosin staining. The combination of the three drugs was found to suppress immune system and decrease the numbers of white blood cells, including lymphocytes. At the eighth week, tumours with a dimension of approximately 1400 mm3 developed. Large atypical nuclei with scant cytoplasm were found to exist using histopathological analysis. No metastasis was observed in the tumour-bearing mice. A combination of CsA, ketoconazole and cyclophosphamide can be used to suppress the immune system in BALB/c mice and induce tumours with significant size.


Asunto(s)
Cetoconazol , Neoplasias , Humanos , Animales , Ratones , Cetoconazol/farmacología , Ratones Endogámicos BALB C , Ratones Desnudos , Ciclofosfamida/farmacología , Ciclosporina , Neoplasias/tratamiento farmacológico
20.
Circulation ; 145(1): 8-17, 2022 01 04.
Artículo en Inglés | MEDLINE | ID: mdl-34503349

RESUMEN

BACKGROUND: Doxycycline was demonstrated in a retrospective study to be associated with greater survival in patients with light chain amyloidosis. Therefore, we prospectively compared the efficacy of bortezomib-cyclophosphamide-dexamethasone (CyBorD) and CyBorD combined with doxycycline for cardiac light chain amyloidosis. METHODS: This was a multicenter, open-label, randomized controlled trial. Patients with Mayo 2004 stage II to III light chain amyloidosis were included. Patients were randomized to doxycycline 100 mg twice daily along with 9 cycles of CyBorD (doxycycline group) or to 9 cycles of CyBorD alone (control group). The primary outcome was 2-year progression-free survival (PFS). PFS was defined as the time from randomization to death, hematologic progression, or organ progression (heart, kidney or liver). Hematologic progression was defined on the basis of a substantial increase in free light chain. An increase in either NT-proBNP (N-terminal pro B-type natriuretic peptide) or cardiac troponin was the main criterion for defining cardiac progression. Cardiac PFS, defined as the time from randomization to cardiac progression or death, was compared between groups in an exploratory analysis. The corresponding treatment hazard ratio was estimated with a Cox regression model. RESULTS: One hundred forty patients underwent randomization, with 70 in each group. The median age was 61 years (range, 33-78 years) with a male:female ratio of 1.75:1. Stage II disease was present in 34 (48.6%) and 33 (47.1%) patients in the doxycycline and control groups, respectively. After a median follow-up duration of 24.4 months, 32 of 70 (45.7%) patients in the doxycycline group and 30 of 70 (42.9%) patients in the control group experienced progression. PFS was not significantly different between groups (hazard ratio, 0.97 [95% CI, 0.59-1.60]; P=0.91). Cardiac progression occurred in 29 of 70 (41.4%) patients in the doxycycline group and 26 of 70 (37.1%) patients in the control group. The death rates for both groups by the end of follow-up was the same, 25 of 70 (35.7%). No significant differences were observed for either cardiac PFS (hazard ratio, 0.91 [95% CI, 0.54-1.55]; P=0.74) or overall survival (hazard ratio, 1.04 [95% CI, 0.60-1.81]; P=0.89). CONCLUSIONS: Our trial demonstrated that doxycycline combined with CyBorD failed to prolong PFS or cardiac PFS compared with CyBorD alone in cardiac light chain amyloidosis. Registration: URL: https://www.clinicaltrials.gov; Unique identifier: NCT03401372.


Asunto(s)
Amiloidosis/tratamiento farmacológico , Bortezomib/uso terapéutico , Ciclofosfamida/uso terapéutico , Dexametasona/uso terapéutico , Doxiciclina/uso terapéutico , Adulto , Anciano , Amiloidosis/psicología , Bortezomib/farmacología , Ciclofosfamida/farmacología , Dexametasona/farmacología , Doxiciclina/farmacología , Femenino , Humanos , Masculino , Persona de Mediana Edad , Estudios Retrospectivos
SELECCIÓN DE REFERENCIAS
Detalles de la búsqueda