Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 10.655
Filter
1.
Sci Rep ; 14(1): 12613, 2024 06 01.
Article in English | MEDLINE | ID: mdl-38824206

ABSTRACT

The aim of the study was to assess healthy tissue metabolism (HTM) using 2-deoxy-2-[18F]fluoro-D-glucose ([18F]FDG) positron emission tomography/computed tomography (PET/CT) during chemotherapy in Hodgkin lymphoma (HL) and the association of HTM with baseline metabolic tumour volume (MTV), haematological parameters, adverse events (AEs), early response and progression-free survival (PFS). We retrospectively identified 200 patients with advanced HL from the RATHL trial with [18F]FDG-PET/CT before (PET0) and following 2 cycles of chemotherapy (PET2). [18F]FDG-uptake was measured in bone marrow (BM), spleen, liver and mediastinal blood pool (MBP). Deauville score (DS) 1-3 was used to classify responders and DS 4-5, non-responders. [18F]FDG-uptake decreased significantly in BM and spleen and increased in liver and MBP at PET2 (all p < 0.0001), but was not associated with MTV. Higher BM uptake at PET0 was associated with lower baseline haemoglobin and higher absolute neutrophil counts, platelets, and white blood cells. High BM, spleen, and liver uptake at PET0 was associated with neutropenia after cycles 1-2. BM uptake at PET0 was associated with treatment failure at PET2 and non-responders with higher BM uptake at PET2 had significantly inferior PFS (p = 0.023; hazard ratio = 2.31). Based on these results, we concluded that the change in HTM during chemotherapy was most likely a direct impact of chemotherapy rather than a change in MTV. BM uptake has prognostic value in HL.


Subject(s)
Fluorodeoxyglucose F18 , Hodgkin Disease , Positron Emission Tomography Computed Tomography , Humans , Hodgkin Disease/drug therapy , Hodgkin Disease/diagnostic imaging , Hodgkin Disease/metabolism , Hodgkin Disease/pathology , Positron Emission Tomography Computed Tomography/methods , Male , Female , Adult , Middle Aged , Prognosis , Retrospective Studies , Young Adult , Bone Marrow/diagnostic imaging , Bone Marrow/metabolism , Bone Marrow/pathology , Bone Marrow/drug effects , Aged , Liver/diagnostic imaging , Liver/metabolism , Liver/pathology , Adolescent , Radiopharmaceuticals , Spleen/diagnostic imaging , Spleen/metabolism , Spleen/pathology
2.
J Toxicol Environ Health A ; 87(16): 647-661, 2024 Aug 17.
Article in English | MEDLINE | ID: mdl-38804873

ABSTRACT

The present study aimed to determine the genoprotective activity and safety of Moringa oleifera leave and Tinospora cordifolia stem extracts against cyclophosphamide (CP)-induced genotoxicity utilizing Swiss albino mice. Animals were divided into 14 groups for subacute treatment with either M. oleifera or T. cordifolia extracts daily for 28 days. The extract doses selected were 100, 200 or 400 mg/kg b.w administered orally alone or combined with CP (50 mg/kg b.w. intraperitoneally daily for 5 days). Analyses performed included the comet assay, micronucleus test (MN) in bone marrow cells and sperm head abnormality assay (SHA). M. oleifera and T. cordifolia extracts induced no significant genotoxic effects on somatic and germ cells. In contrast, for all cells examined M. oleifera and T. cordifolia extracts inhibited DNA damage initiated by CP. Taken together data demonstrated that both plant extracts did not exhibit marked genotoxic effects but displayed potential chemoprotective properties against CP-induced genotoxicity in Swiss mice.


Subject(s)
Cyclophosphamide , DNA Damage , Micronucleus Tests , Moringa oleifera , Plant Extracts , Plant Leaves , Tinospora , Animals , Tinospora/chemistry , Mice , Cyclophosphamide/toxicity , Moringa oleifera/chemistry , Plant Extracts/pharmacology , Male , Plant Leaves/chemistry , DNA Damage/drug effects , Comet Assay , Plant Stems/chemistry , Bone Marrow/drug effects , Bone Marrow Cells/drug effects , Mutagens/toxicity , Antimutagenic Agents/pharmacology
3.
Rev Peru Med Exp Salud Publica ; 41(1): 28-36, 2024 May 27.
Article in Spanish, English | MEDLINE | ID: mdl-38808841

ABSTRACT

OBJECTIVE.: Motivation for the study. Most research supports a negative association between metabolic syndrome and bone health, although there is an overall lack of consensus. Therefore, there is a need for research in this area to develop a better understanding. Main findings. Metabolic syndrome induced by a fructose-rich diet increases the adipogenic predisposition of bone marrow progenitor cells and femoral medullary adiposity in rats. Furthermore, this can be partially prevented by co-treatment with metformin. Implications. Experimental metabolic syndrome has negative effects on bone tissue and can be prevented by oral treatment with metformin as a normoglycemic drug. To determine the effect of metformin (MET) treatment on adipogenic predisposition of bone marrow progenitor cells (BMPC), bone marrow adiposity and bone biomechanical properties. MATERIALS AND METHODS.: 20 young adult male Wistar rats were sorted into four groups. Each of the groups received the following in drinking water: 100% water (C); 20% fructose (F); metformin 100 mg/kg wt/day (M); or fructose plus metformin (FM). After five weeks the animals were sacrificed. Both humeri were dissected to obtain BMPC, and both femurs were dissected to evaluate medullary adiposity (histomorphometry) and biomechanical properties (3-point bending). BMPC were cultured in vitro in adipogenic medium to evaluate RUNX2, PPAR-γ and RAGE expression by RT-PCR, lipase activity and triglyceride accumulation. RESULTS.: The fructose-rich diet (group F) caused an increase in both triglycerides in vitro, and medullary adiposity in vivo; being partially or totally prevented by co-treatment with metformin (group FM). No differences were found in femoral biomechanical tests in vivo, nor in lipase activity and RUNX2/PPAR-γ ratio in vitro. DRF increased RAGE expression in BMPC, being prevented by co-treatment with MET. CONCLUSIONS.: Metabolic syndrome induced by a fructose-rich diet increases femoral medullary adiposity and, in part, the adipogenic predisposition of BMPC. In turn, this can be totally or partially prevented by oral co-treatment with MET.


OBJETIVO.: Motivación para realizar el estudio. La mayoría de las investigaciones respaldan una asociación negativa entre el síndrome metabólico y la salud ósea, aunque existe una falta de consenso general. Por lo tanto, es necesario realizar investigaciones en esta área que permitan desarrollar un mejor conocimiento. Principales hallazgos. El síndrome metabólico inducido por una dieta rica en fructosa incrementa la predisposición adipogénica de células progenitoras de médula ósea y la adiposidad medular femoral en ratas. Además, esto puede prevenirse parcialmente mediante un co-tratamiento con metformina. Implicancias. El síndrome metabólico experimental posee efectos negativos sobre el tejido óseo, pudiendo ser prevenidos mediante un tratamiento oral de metformina como fármaco normoglucemiante. Determinar el efecto de un tratamiento con metformina (MET) sobre la predisposición adipogénica de células progenitoras de médula ósea (CPMO), adiposidad de la médula ósea y propiedades biomecánicas óseas. MATERIALES Y MÉTODOS.: 20 ratas Wistar machos adultos jóvenes fueron separados en cuatro grupos, recibiendo en agua de bebida: 100% agua (C); 20% de fructosa (F); metformina 100 mg/kg peso/día (M); o fructosa más metformina (FM). Tras cinco semanas se sacrificaron los animales, se diseccionaron ambos húmeros para obtener CPMO, y ambos fémures para evaluar adiposidad medular (histomorfometría) y propiedades biomecánicas (flexión a 3 puntos). Las CPMO se cultivaron in vitro en medio adipogénico para evaluar expresión de RUNX2, PPAR-γ y RAGE por RT-PCR, actividad de lipasa y acumulación de triglicéridos. RESULTADOS.: La dieta rica en fructosa (grupo F) produjo un aumento tanto de triglicéridos in vitro, como de la adiposidad medular in vivo; siendo parcial o totalmente prevenido por un co-tratamiento con metformina (grupo FM). No se observaron diferencias en las pruebas biomecánicas femorales in vivo, ni en actividad de lipasa y relación RUNX2/PPAR-γ in vitro. La DRF aumentó la expresión de RAGE en CPMO, siendo prevenido por co-tratamiento con MET. CONCLUSIONES.: El síndrome metabólico inducido por una dieta rica en fructosa aumenta la adiposidad medular femoral y, en parte, la predisposición adipogénica de las CPMO. A su vez, esto puede ser prevenido total o parcialmente por un co-tratamiento oral con MET.


Subject(s)
Adiposity , Femur , Metabolic Syndrome , Metformin , Rats, Wistar , Animals , Metformin/pharmacology , Metabolic Syndrome/etiology , Male , Rats , Adiposity/drug effects , Femur/drug effects , Bone Marrow/drug effects , Hypoglycemic Agents/pharmacology
4.
BMC Endocr Disord ; 24(1): 55, 2024 Apr 28.
Article in English | MEDLINE | ID: mdl-38679740

ABSTRACT

BACKGROUND: Glucocorticoids and sclerostin act as inhibitors of the Wnt signaling pathway, thereby hindering bone formation. Given the pathway's intricate association with mesenchymal stem cells, the hypothesis suggests that heightened sclerostin levels may be intricately linked to an augmentation in marrow adiposity induced by glucocorticoids. This study endeavored to delve into the nuanced relationship between circulating sclerostin and bone marrow adipose tissue in postmenopausal women grappling with glucocorticoid-induced osteoporosis (GIO). METHODS: In this cross-sectional study, 103 patients with autoimmune-associated diseases underwent glucocorticoid treatment, boasting an average age of 61.3 years (standard deviation 7.1 years). The investigation encompassed a thorough assessment, incorporating medical history, anthropometric data, biochemical analysis, and dual-energy X-ray absorptiometry measurements of lumbar and femoral bone mineral density (BMD). Osteoporosis criteria were established at a T-score of -2.5 or lower. Additionally, MR spectroscopy quantified the vertebral marrow fat fraction. RESULTS: BMD at the femoral neck, total hip, and lumbar spine showcased an inverse correlation with marrow fat fraction (r = -0.511 to - 0.647, P < 0.001). Serum sclerostin levels exhibited a positive correlation with BMD at various skeletal sites (r = 0.476 to 0.589, P < 0.001). A noteworthy correlation emerged between circulating sclerostin and marrow fat fraction at the lumbar spine (r = -0.731, 95% CI, -0.810 to -0.627, P < 0.001). Multivariate analysis brought to light that vertebral marrow fat fraction significantly contributed to sclerostin serum concentrations (standardized regression coefficient ß = 0.462, P < 0.001). Even after adjusting for age, body mass index, physical activity, renal function, BMD, and the duration and doses of glucocorticoid treatment, serum sclerostin levels maintained a significant correlation with marrow fat fraction. CONCLUSIONS: Circulating sclerostin levels exhibited a noteworthy association with marrow adiposity in postmenopausal women grappling with GIO.


Subject(s)
Adaptor Proteins, Signal Transducing , Adiposity , Bone Density , Bone Marrow , Glucocorticoids , Postmenopause , Humans , Female , Middle Aged , Glucocorticoids/adverse effects , Cross-Sectional Studies , Adiposity/drug effects , Bone Density/drug effects , Bone Marrow/drug effects , Bone Marrow/metabolism , Aged , Genetic Markers , Biomarkers/blood , Biomarkers/analysis , Osteoporosis, Postmenopausal/blood , Absorptiometry, Photon
5.
Eur J Haematol ; 113(1): 110-116, 2024 Jul.
Article in English | MEDLINE | ID: mdl-38566462

ABSTRACT

OBJECTIVES: Based on a previous phase 1 study, total marrow irradiation (TMI) at 9Gy was added to a myeloablative FluBu4 conditioning regimen in allogeneic hematopoietic stem cell transplantation (HSCT) for myeloid malignancies. Here, we report on the long-term toxicity of TMI combined with FluBu4 and compare it to patients who received only FluBu4. METHODS: We retrospectively analyzed 38 consecutive patients conditioned with FluBu4/TMI (n = 15) or FluBu4 (n = 23, control group) who had at least 1 year follow-up post-transplant. The rate of long-term adverse events that have been previously associated with total body irradiation (TBI) was analyzed in the two groups. RESULTS: The baseline characteristics did not differ between the two groups. The control group had a longer median follow-up (71.2 mo) than the TMI group (38.5 mo) (p = .004). The most common adverse events were xerostomia, dental complications, cataracts, or osteopenia and did not differ between the two groups. Cognitive dysfunction or noninfectious pneumonitis, often detected after high dose TBI, were also not different in the two groups (p = .12 and p = .7, respectively). There was no grade 4 adverse event. CONCLUSION: Our results suggest that a conditioning regimen with TMI 9Gy and FluBu4 does not increase long-term adverse events after allogeneic HSCT.


Subject(s)
Busulfan , Hematopoietic Stem Cell Transplantation , Myeloablative Agonists , Transplantation Conditioning , Transplantation, Homologous , Vidarabine , Humans , Transplantation Conditioning/adverse effects , Transplantation Conditioning/methods , Hematopoietic Stem Cell Transplantation/adverse effects , Hematopoietic Stem Cell Transplantation/methods , Male , Female , Middle Aged , Adult , Vidarabine/analogs & derivatives , Vidarabine/administration & dosage , Vidarabine/adverse effects , Busulfan/adverse effects , Busulfan/administration & dosage , Retrospective Studies , Myeloablative Agonists/adverse effects , Myeloablative Agonists/therapeutic use , Myeloablative Agonists/administration & dosage , Whole-Body Irradiation/adverse effects , Young Adult , Follow-Up Studies , Bone Marrow/radiation effects , Bone Marrow/drug effects , Aged , Adolescent
6.
Stem Cell Res Ther ; 15(1): 123, 2024 Apr 29.
Article in English | MEDLINE | ID: mdl-38679747

ABSTRACT

BACKGROUND: Acute radiation syndrome (ARS) manifests after exposure to high doses of radiation in the instances of radiologic accidents or incidents. Facilitating regeneration of the bone marrow (BM), namely the hematopoietic stem and progenitor cells (HSPCs), is key in mitigating ARS and multi-organ failure. JNJ-26366821, a PEGylated thrombopoietin mimetic (TPOm) peptide, has been shown as an effective medical countermeasure (MCM) to treat hematopoietic-ARS (H-ARS) in mice. However, the activity of TPOm on regulating BM vascular and stromal niches to support HSPC regeneration has yet to be elucidated. METHODS: C57BL/6J mice (9-14 weeks old) received sublethal or lethal total body irradiation (TBI), a model for H-ARS, by 137Cs or X-rays. At 24 h post-irradiation, mice were subcutaneously injected with a single dose of TPOm (0.3 mg/kg or 1.0 mg/kg) or PBS (vehicle). At homeostasis and on days 4, 7, 10, 14, 18, and 21 post-TBI with and without TPOm treatment, BM was harvested for histology, BM flow cytometry of HSPCs, endothelial (EC) and mesenchymal stromal cells (MSC), and whole-mount confocal microscopy. For survival, irradiated mice were monitored and weighed for 30 days. Lastly, BM triple negative cells (TNC; CD45-, TER-119-, CD31-) were sorted for single-cell RNA-sequencing to examine transcriptomics after TBI with or without TPOm treatment. RESULTS: At homeostasis, TPOm expanded the number of circulating platelets and HSPCs, ECs, and MSCs in the BM. Following sublethal TBI, TPOm improved BM architecture and promoted recovery of HSPCs, ECs, and MSCs. Furthermore, TPOm elevated VEGF-C levels in normal and irradiated mice. Following lethal irradiation, mice improved body weight recovery and 30-day survival when treated with TPOm after 137Cs and X-ray exposure. Additionally, TPOm reduced vascular dilation and permeability. Finally, single-cell RNA-seq analysis indicated that TPOm increased the expression of collagens in MSCs to enhance their interaction with other progenitors in BM and upregulated the regeneration pathway in MSCs. CONCLUSIONS: TPOm interacts with BM vascular and stromal niches to locally support hematopoietic reconstitution and systemically improve survival in mice after TBI. Therefore, this work warrants the development of TPOm as a potent radiation MCM for the treatment of ARS.


Subject(s)
Acute Radiation Syndrome , Bone Marrow , Mice, Inbred C57BL , Thrombopoietin , Animals , Male , Mice , Acute Radiation Syndrome/drug therapy , Acute Radiation Syndrome/pathology , Bone Marrow/drug effects , Bone Marrow/radiation effects , Bone Marrow/metabolism , Hematopoietic Stem Cells/drug effects , Hematopoietic Stem Cells/metabolism , Hematopoietic Stem Cells/radiation effects , Stem Cell Niche/drug effects , Stem Cell Niche/radiation effects , Thrombopoietin/pharmacology , Whole-Body Irradiation , Biomimetic Materials/pharmacology , Biomimetic Materials/therapeutic use
7.
Exp Hematol ; 133: 104212, 2024 May.
Article in English | MEDLINE | ID: mdl-38552942

ABSTRACT

Extensive research over the past 50 years has resulted in significant improvements in survival for patients diagnosed with leukemia. Despite this, a subgroup of patients harboring high-risk genetic alterations still suffer from poor outcomes. There is a desperate need for new treatments to improve survival, yet consistent failure exists in the translation of in vitro drug development to clinical application. Preclinical screening conventionally utilizes tumor cell monocultures to assess drug activity; however, emerging research has acknowledged the vital role of the tumor microenvironment in treatment resistance and disease relapse. Current co-culture drug screening methods frequently employ fibroblasts as the designated stromal cell component. Alternative stromal cell types that are known to contribute to chemoresistance are often absent in preclinical evaluations of drug efficacy. This review highlights mechanisms of chemoresistance by a range of different stromal constituents present in the bone marrow microenvironment. Utilizing an array of stromal cell types at the early stages of drug screening may enhance the translation of in vitro drug development to clinical use. Ultimately, we highlight the need to consider the bone marrow microenvironment in drug screening platforms for leukemia to develop superior therapies for the treatment of high-risk patients with poor prognostic outcomes.


Subject(s)
Leukemia , Tumor Microenvironment , Humans , Tumor Microenvironment/drug effects , Leukemia/pathology , Leukemia/drug therapy , Drug Screening Assays, Antitumor/methods , Antineoplastic Agents/pharmacology , Antineoplastic Agents/therapeutic use , Drug Resistance, Neoplasm , Animals , Bone Marrow/pathology , Bone Marrow/drug effects , Bone Marrow/metabolism , Stromal Cells/pathology , Stromal Cells/metabolism , Stromal Cells/drug effects , Coculture Techniques , Mesenchymal Stem Cells/metabolism , Mesenchymal Stem Cells/drug effects , Mesenchymal Stem Cells/pathology
8.
Nature ; 625(7993): 166-174, 2024 Jan.
Article in English | MEDLINE | ID: mdl-38057662

ABSTRACT

Myeloid cells are known to suppress antitumour immunity1. However, the molecular drivers of immunosuppressive myeloid cell states are not well defined. Here we used single-cell RNA sequencing of human and mouse non-small cell lung cancer (NSCLC) lesions, and found that in both species the type 2 cytokine interleukin-4 (IL-4) was predicted to be the primary driver of the tumour-infiltrating monocyte-derived macrophage phenotype. Using a panel of conditional knockout mice, we found that only deletion of the IL-4 receptor IL-4Rα in early myeloid progenitors in bone marrow reduced tumour burden, whereas deletion of IL-4Rα in downstream mature myeloid cells had no effect. Mechanistically, IL-4 derived from bone marrow basophils and eosinophils acted on granulocyte-monocyte progenitors to transcriptionally programme the development of immunosuppressive tumour-promoting myeloid cells. Consequentially, depletion of basophils profoundly reduced tumour burden and normalized myelopoiesis. We subsequently initiated a clinical trial of the IL-4Rα blocking antibody dupilumab2-5 given in conjunction with PD-1/PD-L1 checkpoint blockade in patients with relapsed or refractory NSCLC who had progressed on PD-1/PD-L1 blockade alone (ClinicalTrials.gov identifier NCT05013450 ). Dupilumab supplementation reduced circulating monocytes, expanded tumour-infiltrating CD8 T cells, and in one out of six patients, drove a near-complete clinical response two months after treatment. Our study defines a central role for IL-4 in controlling immunosuppressive myelopoiesis in cancer, identifies a novel combination therapy for immune checkpoint blockade in humans, and highlights cancer as a systemic malady that requires therapeutic strategies beyond the primary disease site.


Subject(s)
Bone Marrow , Carcinogenesis , Interleukin-4 , Myelopoiesis , Signal Transduction , Animals , Humans , Mice , B7-H1 Antigen/antagonists & inhibitors , B7-H1 Antigen/metabolism , Bone Marrow/drug effects , Bone Marrow/metabolism , Carcinogenesis/drug effects , Carcinogenesis/metabolism , Carcinogenesis/pathology , Carcinoma, Non-Small-Cell Lung/immunology , Carcinoma, Non-Small-Cell Lung/metabolism , Carcinoma, Non-Small-Cell Lung/pathology , Carcinoma, Non-Small-Cell Lung/therapy , CD8-Positive T-Lymphocytes/drug effects , CD8-Positive T-Lymphocytes/immunology , Immune Checkpoint Inhibitors/immunology , Immune Checkpoint Inhibitors/pharmacology , Immune Checkpoint Inhibitors/therapeutic use , Interleukin-4/metabolism , Lung Neoplasms/immunology , Lung Neoplasms/metabolism , Lung Neoplasms/pathology , Lung Neoplasms/therapy , Lymphocytes, Tumor-Infiltrating/drug effects , Lymphocytes, Tumor-Infiltrating/immunology , Monocytes/drug effects , Programmed Cell Death 1 Receptor/antagonists & inhibitors , Programmed Cell Death 1 Receptor/metabolism , Recurrence , Signal Transduction/drug effects
9.
Toxicology ; 492: 153532, 2023 06 15.
Article in English | MEDLINE | ID: mdl-37141935

ABSTRACT

Thallium (Tl) is a high-priority toxic metal that poses a severe threat to human health. The toxicity characteristics induced by Tl have been partially discussed. However, the immunotoxic effects of Tl exposure have remained largely unexplored. Our findings demonstrated that 50 ppm of Tl exposure for one week induced severe weight loss in mice, which was accompanied by appetite suppression. Moreover, although Tl exposure did not induce significant pathological damage to skeletal muscle and bone, Tl inhibited the expression of B cell development-related genes in the bone marrow. Additionally, Tl exposure increased B cell apoptosis and reduced its generation in the bone marrow. Analysis of B cells in the blood indicated that the percentage of B-2 cells decreased significantly, whereas B-2 cell proportions in the spleen did not. The percentage of CD4+ T cells in the thymus increased significantly, and the proportion of CD8+ T cells did not. Furthermore, although the proportion of the total CD4+ and CD8+ T cells was not significantly altered in the blood and spleen, Tl exposure promoted the migration of naïve CD4+ T cells and recent thymic emigrants (RTEs) from the thymus to the spleen. These results suggest that Tl exposure can affect B and T cell generation and migration, which provides new evidence for Tl-induced immunotoxicity.


Subject(s)
B-Lymphocytes , T-Lymphocytes , Thallium , Thallium/toxicity , B-Lymphocytes/cytology , B-Lymphocytes/drug effects , T-Lymphocytes/drug effects , Animals , Mice , Cell Movement/drug effects , Gene Expression/drug effects , CD4-Positive T-Lymphocytes/cytology , CD4-Positive T-Lymphocytes/drug effects , Thymus Gland/cytology , Thymus Gland/drug effects , Bone Marrow/drug effects , Body Weight/drug effects
10.
J Med Case Rep ; 16(1): 294, 2022 Jul 31.
Article in English | MEDLINE | ID: mdl-35907896

ABSTRACT

BACKGROUND: Liver involvement in adults with acute myeloid leukemia is uncommon. Most of the case reports describe acute liver failure or obstructive jaundice, while acute hepatitis is rarely mentioned. We report a patient with acute myeloid leukemia who presented with clinical, biochemical, and radiological signs of acute hepatitis that totally regressed after chemotherapy. CASE PRESENTATION: A 38-year-old Caucasian man presented with fever, cough, and mild fatigue. Laboratory workup showed anemia, thrombocytopenia, severe leukocytosis, transaminitis, and hyperbilirubinemia. Imaging of the abdomen (ultrasound and magnetic resonance) showed hepatomegaly, splenomegaly, upper limits portal veins diameters, increased thickness of the gallbladder wall, and significant abdominal lymph nodes. Peripheral blood smear and bone marrow evaluation were consistent with acute myeloid leukemia, and liver biopsy showed massive sinusoidal and portal infiltration by leukemic cells. After remission-inducing chemotherapy, there was complete normalization of liver function tests, and liver, spleen, and portal vein size. CONCLUSIONS: This case highlights the importance of taking acute myeloid leukemia into account as a possible cause of liver damage to make a rapid diagnosis and start appropriate treatment that may lead to hematological remission and hepatic dysfunction resolution.


Subject(s)
Cholestasis , Core Binding Factor beta Subunit , Hepatitis , Leukemia, Myeloid, Acute , Myosin Heavy Chains , Acute Disease , Adult , Bone Marrow/drug effects , Bone Marrow/pathology , Cholestasis/complications , Cholestasis/drug therapy , Cholestasis/pathology , Hepatitis/complications , Hepatitis/diagnosis , Hepatitis/drug therapy , Humans , Leukemia, Myeloid, Acute/complications , Leukemia, Myeloid, Acute/drug therapy , Liver/drug effects , Liver/pathology , Liver/physiology , Liver/physiopathology , Male
11.
Mar Drugs ; 20(3)2022 Mar 09.
Article in English | MEDLINE | ID: mdl-35323500

ABSTRACT

Hematopoietic damage is a serious side effect of cytotoxic drugs, and agents promoting hematopoiesis are quite important for decreasing the death rate in cancer patients. In our previous work, we prepared the simulated digestive product of fucoidan from Sargassum fusiforme, DSFF, and found that DSFF could activate macrophages. However, more investigations are needed to further evaluate whether DSFF could promote hematopoiesis in the chemotherapy process. In this study, the protective effect of DSFF (1.8-7.2 mg/kg, i.p.) on cyclophosphamide-induced hematopoietic damage in mice and the underlying mechanisms were investigated. Our results show that DSFF could restore the numbers of white blood cells, neutrophils, and platelets in the peripheral blood, and could also retard bone marrow cell decrease in mice with cyclophosphamide-induced hematopoietic damage. UPLC/Q-Extraction Orbitrap/MS/MS-based lipidomics results reveal 16 potential lipid biomarkers in a serum that responded to hematopoietic damage in mice. Among them, PC (20:1/14:0) and SM (18:0/22:0) were the key lipid molecules through which DSFF exerted protective actions. In a validation experiment, DSFF (6.25-100 µg/mL) could also promote K562 cell proliferation and differentiation in vitro. The current findings indicated that DSFF could affect the blood cells and bone marrow cells in vivo and thus showed good potential and application value in alleviating the hematopoietic damage caused by cyclophosphamide.


Subject(s)
Cyclophosphamide/toxicity , Hematopoiesis/drug effects , Myeloablative Agonists/toxicity , Polysaccharides/pharmacology , Protective Agents/pharmacology , Sargassum , Animals , Biomarkers/blood , Bone Marrow/drug effects , Bone Marrow/metabolism , Cell Proliferation/drug effects , DNA/metabolism , Humans , K562 Cells , Leukocyte Count , Lipidomics , Mice , Neutrophils/drug effects , Platelet Count
12.
Toxins (Basel) ; 14(2)2022 01 29.
Article in English | MEDLINE | ID: mdl-35202133

ABSTRACT

The aim of this study was to determine whether low doses of zearalenone (ZEN) influence the carry-over of ZEN and its metabolites to the bone marrow microenvironment and, consequently, haematological parameters. Pre-pubertal gilts (with a body weight of up to 14.5 kg) were exposed to daily ZEN doses of 5 µg/kg BW (group ZEN5, n = 15), 10 µg/kg BW (group ZEN10, n = 15), 15 µg/kg BW (group ZEN15, n = 15), or were administered a placebo (group C, n = 15) throughout the entire experiment. Bone marrow was sampled on three dates (exposure dates 7, 21, and 42-after slaughter) and blood for haematological analyses was sampled on 10 dates. Significant differences in the analysed haematological parameters (WBC White Blood Cells, MONO-Monocytes, NEUT-Neutrophils, LYMPH-Lymphocytes, LUC-Large Unstained Cells, RBC-Red Blood Cells, HGB-Haemoglobin, HCT-Haematocrit, MCH-Mean Corpuscular Volume, MCHC-Mean Corpuscular Haemoglobin Concentrations, PLT-Platelet Count and MPV-Mean Platelet Volume) were observed between groups. The results of the experiment suggest that exposure to low ZEN doses triggered compensatory and adaptive mechanisms, stimulated the local immune system, promoted eryptosis, intensified mycotoxin biotransformation processes in the liver, and produced negative correlations between mycotoxin concentrations and selected haematological parameters.


Subject(s)
Bone Marrow/drug effects , Plasma/drug effects , Zearalenone/toxicity , Animals , Female , Hematologic Tests , No-Observed-Adverse-Effect Level , Sexual Maturation , Swine
13.
Nat Commun ; 13(1): 121, 2022 01 10.
Article in English | MEDLINE | ID: mdl-35013215

ABSTRACT

HIV is difficult to eradicate due to the persistence of a long-lived reservoir of latently infected cells. Previous studies have shown that natural killer cells are important to inhibiting HIV infection, but it is unclear whether the administration of natural killer cells can reduce rebound viremia when anti-retroviral therapy is discontinued. Here we show the administration of allogeneic human peripheral blood natural killer cells delays viral rebound following interruption of anti-retroviral therapy in humanized mice infected with HIV-1. Utilizing genetically barcoded virus technology, we show these natural killer cells efficiently reduced viral clones rebounding from latency. Moreover, a kick and kill strategy comprised of the protein kinase C modulator and latency reversing agent SUW133 and allogeneic human peripheral blood natural killer cells during anti-retroviral therapy eliminated the viral reservoir in a subset of mice. Therefore, combinations utilizing latency reversal agents with targeted cellular killing agents may be an effective approach to eradicating the viral reservoir.


Subject(s)
Anti-HIV Agents/pharmacology , CD4-Positive T-Lymphocytes/immunology , HIV Infections/therapy , HIV-1/drug effects , Killer Cells, Natural/immunology , Protein Kinase Inhibitors/pharmacology , Viremia/therapy , Animals , Bone Marrow/drug effects , Bone Marrow/immunology , Bone Marrow/virology , CD4-Positive T-Lymphocytes/drug effects , CD4-Positive T-Lymphocytes/virology , Coculture Techniques , Female , HIV Infections/genetics , HIV Infections/immunology , HIV Infections/virology , HIV-1/genetics , HIV-1/immunology , Host-Pathogen Interactions/drug effects , Host-Pathogen Interactions/genetics , Host-Pathogen Interactions/immunology , Humans , Killer Cells, Natural/transplantation , Male , Mice , Mice, Transgenic , Protein Kinase C/genetics , Protein Kinase C/immunology , Spleen/drug effects , Spleen/immunology , Spleen/virology , Viral Load/drug effects , Viremia/genetics , Viremia/immunology , Viremia/virology , Virus Latency/drug effects , Virus Replication/drug effects
14.
Blood Cancer J ; 12(1): 19, 2022 01 28.
Article in English | MEDLINE | ID: mdl-35091554

ABSTRACT

Hypomethylating agents (HMA) like azacitidine are licensed for the treatment of acute myeloid leukemia (AML) patients ineligible for allogeneic hematopoietic stem cell transplantation. Biomarker-driven identification of HMA-responsive patients may facilitate the choice of treatment, especially in the challenging subgroup above 60 years of age. Since HMA possesses immunomodulatory functions that constitute part of their anti-tumor effect, we set out to analyze the bone marrow (BM) immune environment by next-generation sequencing of T cell receptor beta (TRB) repertoires in 51 AML patients treated within the RAS-AZIC trial. Patients with elevated pretreatment T cell diversity (11 out of 41 patients) and those with a boost of TRB richness on day 15 after azacitidine treatment (12 out of 46 patients) had longer event-free and overall survival. Both pretreatment and dynamic BM T cell metrics proved to be better predictors of outcome than other established risk factors. The favorable broadening of the BM T cell space appeared to be driven by antigen since these patients showed significant skewing of TRBV gene usage. Our data suggest that one course of AZA can cause reconstitution to a more physiological T cell BM niche and that the T cell space plays an underestimated prognostic role in AML.Trial registration: DRKS identifier: DRKS00004519.


Subject(s)
Antimetabolites, Antineoplastic/therapeutic use , Azacitidine/therapeutic use , Bone Marrow/drug effects , Leukemia, Myeloid, Acute/drug therapy , T-Lymphocytes/drug effects , Aged , Aged, 80 and over , Bone Marrow/pathology , Female , Humans , Leukemia, Myeloid, Acute/epidemiology , Leukemia, Myeloid, Acute/pathology , Male , Middle Aged , Survival Analysis , T-Lymphocytes/pathology
15.
Leukemia ; 36(2): 333-347, 2022 02.
Article in English | MEDLINE | ID: mdl-34518644

ABSTRACT

We show that pro-inflammatory oncostatin M (OSM) is an important regulator of hematopoietic stem cell (HSC) niches in the bone marrow (BM). Treatment of healthy humans and mice with granulocyte colony-stimulating factor (G-CSF) dramatically increases OSM release in blood and BM. Using mice null for the OSM receptor (OSMR) gene, we demonstrate that OSM provides a negative feed-back acting as a brake on HSPC mobilization in response to clinically relevant mobilizing molecules G-CSF and CXCR4 antagonist. Likewise, injection of a recombinant OSM molecular trap made of OSMR complex extracellular domains enhances HSC mobilization in poor mobilizing C57BL/6 and NOD.Cg-PrkdcscidIl2rgtm1Wjl/SzJ mice. Mechanistically, OSM attenuates HSC chemotactic response to CXCL12 and increases HSC homing to the BM signaling indirectly via BM endothelial and mesenchymal cells which are the only cells expressing OSMR in the BM. OSM up-regulates E-selectin expression on BM endothelial cells indirectly increasing HSC proliferation. RNA sequencing of HSCs from Osmr-/- and wild-type mice suggest that HSCs have altered cytoskeleton reorganization, energy usage and cycling in the absence of OSM signaling in niches. Therefore OSM is an important regulator of HSC niche function restraining HSC mobilization and anti-OSM therapy combined with current mobilizing regimens may improve HSPC mobilization for transplantation.


Subject(s)
Bone Marrow/physiology , Granulocyte Colony-Stimulating Factor/administration & dosage , Hematopoietic Stem Cell Mobilization/methods , Hematopoietic Stem Cells/cytology , Oncostatin M/metabolism , Stem Cell Niche , Animals , Bone Marrow/drug effects , Female , Hematopoietic Stem Cells/drug effects , Hematopoietic Stem Cells/metabolism , Humans , Male , Mice , Mice, Inbred C57BL , Mice, Inbred NOD
16.
Acta Radiol ; 63(2): 245-252, 2022 Feb.
Article in English | MEDLINE | ID: mdl-33497273

ABSTRACT

BACKGROUND: Polyphenols in extra virgin olive oil (EVOO) have been found to reduce the expression of PPARγ2, inhibit adipocyte differentiation, and enhance the formation of osteoblasts from bone marrow stem cells. However, the underlying mechanisms of their action remain unknown. PURPOSE: To determine the sequential effects of EVOO on marrow fat expansion induced by estrogen deprivation using 3.0-T proton magnetic resonance (MR) spectroscopy in an ovariectomy (OVX) rabbit model of postmenopausal bone loss over a six-month period. MATERIAL AND METHODS: A total of 45 female New Zealand rabbits were equally divided into sham-operation, OVX controls, and OVX treated with EVOO for six months. Marrow fat fraction was measured by MR spectroscopy at baseline conditions, and three and six months postoperatively, respectively. Serum bone biomarkers, lumbar and femoral bone mineral density, microtomographic parameters, biomechanical properties, and quantitative parameters of marrow adipocytes were studied. RESULTS: OVX was associated with marrow adiposity in a time-dependent manner, accompanied with increased bone turnover and impaired bone mass and trabecular microarchitecture. In OVX rabbits, EVOO markedly alleviated trabecular bone loss and reduced the accumulation of lipid droplets including adipocyte size, density, and areas of fat deposits in the bone marrow. EVOO prevented such changes in terms of both marrow adiposity and bone remodeling. CONCLUSION: Early EVOO treatment may exert beneficial effects on bone by modulating marrow adiposity, which would support their protective effect against bone pathologies.


Subject(s)
Adiposity/drug effects , Bone Marrow/drug effects , Olive Oil/pharmacology , Osteoporosis, Postmenopausal/physiopathology , PPAR gamma/antagonists & inhibitors , Polyphenols/pharmacology , Proton Magnetic Resonance Spectroscopy , Adiposity/physiology , Animals , Biomechanical Phenomena , Bone Density/drug effects , Bone Marrow/physiology , Bone Marrow Cells/cytology , Disease Models, Animal , Female , Humans , Osteogenesis , Ovariectomy , Rabbits
17.
Endocrinology ; 163(2)2022 02 01.
Article in English | MEDLINE | ID: mdl-34864986

ABSTRACT

Glucocorticoids (GCs) are critical modulators of the immune system. The hypothalamic-pituitary-adrenal (HPA) axis regulates circulating GC levels and is stimulated by endotoxins. Lymphoid organs also produce GCs; however, it is not known how lymphoid GC levels are regulated in response to endotoxins. We assessed whether an acute challenge of lipopolysaccharide (LPS) increases lymphoid levels of progesterone and GCs, and expression of steroidogenic enzymes and key HPA axis components (eg, corticotropin-releasing hormone [CRH], adrenocorticotropic hormone [ACTH]). We administered LPS (50 µg/kg intraperitoneally) or vehicle control to male and female C57BL/6J neonatal (postnatal day [PND] 5) and adult (PND90) mice and collected blood, bone marrow, thymus, and spleen 4 hours later. We measured progesterone, 11-deoxycorticosterone, corticosterone, and 11-dehydrocorticosterone via liquid chromatography-tandem mass spectrometry. We measured gene expression of key steroidogenic enzymes (Cyp11b1, Hsd11b1, and Hsd11b2) and HPA axis components (Crh, Crhr1, Pomc, and Mc2r) via quantitative polymerase chain reaction. At PND5, LPS induced greater increases in steroid levels in lymphoid organs than in blood. In contrast, at PND90, LPS induced greater increases in steroid levels in blood than in lymphoid organs. Steroidogenic enzyme transcripts were present in all lymphoid organs, and LPS altered steroidogenic enzyme expression predominantly in the spleen. Lastly, we detected transcripts of key HPA axis components in all lymphoid organs, and there was an effect of LPS in the spleen. Taken together, these data suggest that LPS regulates GC production by lymphoid organs, similar to its effects on the adrenal glands, and the effects of LPS might be mediated by local expression of CRH and ACTH.


Subject(s)
Bone Marrow/metabolism , Glucocorticoids/biosynthesis , Lipopolysaccharides/pharmacology , Spleen/metabolism , Thymus Gland/metabolism , 11-beta-Hydroxysteroid Dehydrogenase Type 1/genetics , 11-beta-Hydroxysteroid Dehydrogenase Type 2/genetics , Animals , Animals, Newborn/metabolism , Bone Marrow/drug effects , Bone Marrow/enzymology , Corticosterone/analysis , Corticosterone/blood , Female , Glucocorticoids/blood , Hypothalamo-Hypophyseal System/drug effects , Immunity, Innate/drug effects , Male , Mice , Mice, Inbred C57BL , Pituitary-Adrenal System/drug effects , RNA, Messenger/analysis , Receptors, Corticotropin-Releasing Hormone/genetics , Spleen/drug effects , Spleen/enzymology , Steroid 11-beta-Hydroxylase/genetics , Thymus Gland/drug effects , Thymus Gland/enzymology
19.
Clin Breast Cancer ; 22(1): e1-e7, 2022 01.
Article in English | MEDLINE | ID: mdl-34078565

ABSTRACT

BACKGROUND: The coronavirus disease 2019 pandemic is a global public health event. Wuhan used to be the epicenter of China and finally controlled the outbreak through city lockdown and many other policies. However, the pandemic and the prevention strategies had a huge impact on the medical care procedures for patients with breast cancer, leading to the delay or interruption of anticancer therapies. PATIENTS AND METHODS: To better serve patients with breast cancer under the premise of epidemic control, many strategies have been proposed and optimized in our center. One of the most important parts of these strategies is the promotion of telemedicine, including online consultation, online prescription, and drug mailing services. RESULTS: In keeping with the city and hospital policies, we have also introduced stricter ward management policies and more precise care. CONCLUSION: Here, we collected the diagnosis and treatment process of patients with breast cancer in our center during the coronavirus disease 2019 pandemic, which was found to be correlated to a reduction in chemotherapy-related myelosuppression and hepatic dysfunction, hoping to provide a reference for other cancer centers that may suffer from the similar situation.


Subject(s)
Breast Neoplasms/drug therapy , COVID-19/epidemiology , SARS-CoV-2 , Adult , Aged , Antineoplastic Agents/adverse effects , Bone Marrow/drug effects , Breast Neoplasms/diagnosis , Breast Neoplasms/pathology , Chemical and Drug Induced Liver Injury/etiology , China/epidemiology , Female , Humans , Middle Aged , Telemedicine
20.
J Histochem Cytochem ; 70(2): 169-179, 2022 02.
Article in English | MEDLINE | ID: mdl-34915746

ABSTRACT

Osteoclasts are cells whose main function is the resorption of bone matrix. However, several factors, including medications, can interfere with the resorption process. Alendronate (ALN), a nitrogen-containing type of bisphosphonate, and dexamethasone (DEX), a glucocorticoid, are drugs that may affect the resorption activity. The aim of this study is to investigate the effects of ALN, and/or DEX on osteoclast gene expression and resorption activity in primary mouse marrow cultures stimulated with 1,25-dihydroxyvitamin D3, a model for the bone microenvironment. Cultures were treated only with ALN (10-5 M), DEX (10-6 M), and with a combination of both agents. Viability assays performed at days 5, 7, and 9 showed the highest number of viable cells at day 7. All the following assays were then performed at day 7 of cell culture: tartrate resistant acid phosphatase (TRAP) histochemistry, receptor activator of nuclear factor kappa B ligand (RANKL) immunofluorescence, osteoprotegerin (OPG), and RANKL gene expression by qPCR and resorption analysis by scanning electron microscopy. Treatment with ALN, DEX, and the combination of both did not promote significant changes in the number of TRAP+ cells, although larger giant cells were detected in groups treated with DEX. DEX treatment increased the gene expression of RANKL and reduced OPG. The treatment with ALN reduced the depth of the resorption pits, but their inhibitory effect was less effective when administered with DEX.


Subject(s)
Alendronate/pharmacology , Bone Marrow/drug effects , Bone Resorption/drug therapy , Dexamethasone/pharmacology , Osteoclasts/drug effects , Animals , Cattle , Cell Survival/drug effects , Cells, Cultured , Dose-Response Relationship, Drug , Mice , Mice, Inbred BALB C
SELECTION OF CITATIONS
SEARCH DETAIL
...