Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 312
Filtrar
Más filtros

Banco de datos
Tipo del documento
Intervalo de año de publicación
1.
Genes Chromosomes Cancer ; 62(7): 377-391, 2023 07.
Artículo en Inglés | MEDLINE | ID: mdl-36562080

RESUMEN

Small cell lung cancer (SCLC) is treated as a homogeneous disease, although the expression of NEUROD1, ASCL1, POU2F3, and YAP1 identifies distinct molecular subtypes. The MYC oncogene, amplified in SCLC, was recently shown to act as a lineage-specific factor to associate subtypes with histological classes. Indeed, MYC-driven SCLCs show a distinct metabolic profile and drug sensitivity. To disentangle their molecular features, we focused on the co-amplified PVT1, frequently overexpressed and originating circular (circRNA) and chimeric RNAs. We analyzed hsa_circ_0001821 (circPVT1) and PVT1/AKT3 (chimPVT1) as examples of such transcripts, respectively, to unveil their tumorigenic contribution to SCLC. In detail, circPVT1 activated a pro-proliferative and anti-apoptotic program when over-expressed in lung cells, and knockdown of chimPVT1 induced a decrease in cell growth and an increase of apoptosis in SCLC in vitro. Moreover, the investigated PVT1 transcripts underlined a functional connection between MYC and YAP1/POU2F3, suggesting that they contribute to the transcriptional landscape associated with MYC amplification. In conclusion, we have uncovered a functional role of circular and chimeric PVT1 transcripts in SCLC; these entities may prove useful as novel biomarkers in MYC-amplified tumors.


Asunto(s)
Neoplasias Pulmonares , Carcinoma Pulmonar de Células Pequeñas , Humanos , Carcinoma Pulmonar de Células Pequeñas/genética , Neoplasias Pulmonares/genética , Proliferación Celular/genética , Apoptosis/genética , Línea Celular Tumoral , Regulación Neoplásica de la Expresión Génica , Proteínas Proto-Oncogénicas c-akt/genética
2.
Eur J Immunol ; 52(7): 1171-1189, 2022 07.
Artículo en Inglés | MEDLINE | ID: mdl-35562849

RESUMEN

Common variable immunodeficiency (CVID) is the most frequent primary antibody deficiency whereby follicular helper T (Tfh) cells fail to establish productive responses with B cells in germinal centers. Here, we analyzed the frequency, phenotype, transcriptome, and function of circulating Tfh (cTfh) cells in CVID patients displaying autoimmunity as an additional phenotype. A group of patients showed a high frequency of cTfh1 cells and a prominent expression of PD-1 and ICOS as well as a cTfh mRNA signature consistent with highly activated, but exhausted, senescent, and apoptotic cells. Plasmatic CXCL13 levels were elevated in this group and positively correlated with cTfh1 cell frequency and PD-1 levels. Monoallelic variants in RTEL1, a telomere length- and DNA repair-related gene, were identified in four patients belonging to this group. Their blood lymphocytes showed shortened telomeres, while their cTfh were more prone to apoptosis. These data point toward a novel pathogenetic mechanism in CVID, whereby alterations in DNA repair and telomere elongation might predispose to antibody deficiency. A Th1, highly activated but exhausted and apoptotic cTfh phenotype was associated with this form of CVID.


Asunto(s)
Inmunodeficiencia Variable Común , Apoptosis/genética , Inmunodeficiencia Variable Común/genética , Humanos , Receptor de Muerte Celular Programada 1/genética , Células T Auxiliares Foliculares , Linfocitos T Colaboradores-Inductores
3.
Eur J Clin Invest ; 53(2): e13898, 2023 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-36380693

RESUMEN

BACKGROUND: Controversies on sub-populations most sensitive to therapy and the best timing of starting the treatment still surround the use of immunomodulatory drugs in COVID-19. OBJECTIVES: We designed a multicentre open-label randomised controlled trial to test the effect of prompt adding of tofacitinib to standard therapy for hospitalised patients affected by mild/moderate COVID-19 pneumonitis. METHODS: Patients admitted to three Italian hospitals affected by COVID-19 pneumonitis not requiring mechanical ventilation were randomised to receive standard treatment alone or tofacitinib (10 mg/bid) for 2 weeks, starting within the first 24 h from admission. RESULTS: A total of 116 patients were randomised; 49 in the experimental arm completed the 14-day treatment period, 9 discontinued tofacitinib as the disease worsened and were included in the analysis, and 1 died of respiratory failure. All 58 control patients completed the study. Clinical and demographic characteristics were similar between the study groups. In the tofacitinib group, 9/58 (15.5%) patients progressed to noninvasive ventilation (CPAP) to maintain SO2 > 93%, invasive mechanical ventilation or death by day 14 was 15.5%, significantly less than in the control group (20/58, 34.4%, RR 0,45, RRR -55%, NNT 5; p = .018). No differences in severe adverse effect incidence had been observed across the groups. CONCLUSION: High-dose tofacitinib therapy in patients with COVID pneumonitis is safe and may prevent deterioration to respiratory failure.


Asunto(s)
COVID-19 , Insuficiencia Respiratoria , Humanos , SARS-CoV-2 , Tratamiento Farmacológico de COVID-19 , Resultado del Tratamiento
4.
J Pathol ; 256(4): 402-413, 2022 04.
Artículo en Inglés | MEDLINE | ID: mdl-34919276

RESUMEN

Multiple myeloma (MM) progression and drug resistance depend on the crosstalk between MM cells and bone marrow (BM) fibroblasts (FBs). During monoclonal gammopathy of undetermined significance (MGUS) to MM transition, MM cell-derived exosomes (EXOs) reprogram the miRNA (miR) profile of FBs, inducing the overexpression miR-23b-3p, miR-27b-3p, miR-125b-5p, miR-214-3p, and miR-5100. Here, we demonstrate that the miR content of MM FB-derived EXOs (FB-EXOs) overlaps the miR profile of parental FBs by overexpressing comparable levels of miR-23b-3p, miR-27b-3p, miR-125b-5p, miR-214-3p, and miR-5100. Recipient MM cells co-cultured with MM FB-EXOs selectively overexpress only miR-214-3p and miR-5100 but not miR-23b-3p, miR-27b-3p, and miR-125b-5p, suggesting a putative selective transfer. MM cells express HOTAIR, TOB1-AS1, and MALAT1 lncRNAs. Transient transfection of MM cells with lnc·siRNAs demonstrates that HOTAIR, TOB1-AS1, and MALAT1 lncRNAs are sponges for miR-23b-3p, miR-27b-3p, and miR-125b-5p. Indeed, lncRNA knockdown significantly increased miR levels in U266 MM cells co-cultured with MM FB-EXOs. Selective miR-214-3p and miR-5100 overexpression modulates MAPK, PI3K/AKT/mTOR, and p53 pathways in MM cells. Interrogation using the DIANA tools algorithm and transient overexpression using miR mimic probes confirmed the involvement of miR-214-3p and miR-5100 and their target genes, PTEN and DUSP16, respectively, in the modulation of these intracellular pathways. Finally, the uptake of EXOs as well as miR-214-3p and miR-5100 overexpression increase MM cell proliferation and resistance to bortezomib-induced apoptosis by switching the balance between pro-/anti-apoptotic proteins. Overall, these data show that MM cells are not simply a container into which EXOs empty their cargo. On the contrary, tumour cells finely neutralize exosomal miRs via lncRNA expression to ensure their survival. © 2021 The Pathological Society of Great Britain and Ireland.


Asunto(s)
Exosomas , MicroARNs , Mieloma Múltiple , ARN Largo no Codificante , Exosomas/patología , Fibroblastos/patología , Humanos , MicroARNs/genética , MicroARNs/metabolismo , Mieloma Múltiple/patología , Fosfatidilinositol 3-Quinasas/metabolismo , ARN Largo no Codificante/genética , ARN Largo no Codificante/metabolismo
5.
Adv Exp Med Biol ; 1438: 223-229, 2023.
Artículo en Inglés | MEDLINE | ID: mdl-37845465

RESUMEN

BACKGROUND: Angiotensin Converting Enzyme 2 (ACE2) is an endothelial cell receptor used by SARS-CoV- 2 virus to enter cells. Pulmonary function tests (PFTs), mainly spirometry, are the main diagnostic tools for most respiratory diseases. PFTs are mandatory for assessing the response to therapy. AIM: We evaluated patients after the SARS-CoV-2 infection through flow-volume spirometry that evaluates the role of drugs inhibiting the ACE2 pathway. MATERIAL AND METHODS: We evaluated 112 Caucasian patients 3-6 months after COVID-19 disease, i.e. after the date of negative molecular or antigenic nasopharyngeal swab. The series of patients showed a great variability due to a wide spectrum of age, the severity of disease manifestations, hospitalization, invasive/non-invasive ventilation, comorbidities, the presence/absence of a previous pneumological diagnosis and the variants of the virus. Patients were divided into those who were being treated with angiotensin receptor blocker (ARB) or ACE2 inhibitors (ACEi) (ARB/ACEi, group 1, 23 females and 12 males, aged 63.63 ± 10.40), and those who were not treated with these drugs (group 2, 38 females and 37 males, aged 55.12 ± 16.51). Distal airflow obstruction (DAO) was evaluate as forced expiratory flow (FEF) at 25%, 50% and 75% of total flow. RESULTS: Group 1 presented lower peripheral oxygen saturation percentage vs group 2 (96.54 ± 3.06 vs 97.30 ± 1.19%, p < 0.05). Spirometry data were worst in group1: Forced expiratory volume at first minute (FEV1) (91.20 ± 17.09 vs 97.56 ± 16.40%, p < 0.05), Forced vital capacity (94.06 ± 17.48 vs 99.13 ± 17.71%, p < 0.05), and Tiffenau Index (0.78 ± 0.12 vs 0.84 ± 0.10, p < 0.05). There was a DAO in group1. In group 1, we found also a reduction in FEF 25 (73.97 ± 27.28 vs 86.89 ± 22.44%, p < 0.05), FEF 50 (74.69 ± 33.01 vs 85.67 ± 23.74%, p < 0.05), and FEF 25-75 (74.14 ± 35.03 vs 83.92 ± 25.38%, p < 0.05) but not in FEF 75 (73.06 ± 39.37 vs 82.27 ± 43.33%, p < 0.05). DISCUSSION: In patients treated with ARB/ACEi the indexes of respiratory function were shifted towards the lower limits (albeit within normal limits). These parameters were significantly reduced compared to patients not treated with these drugs. This indicates that the COVID-19 disease is not only a pulmonary disease, but also a vascular one.


Asunto(s)
COVID-19 , Masculino , Femenino , Humanos , COVID-19/diagnóstico , SARS-CoV-2 , Enzima Convertidora de Angiotensina 2 , Angiotensinas , Antagonistas de Receptores de Angiotensina/uso terapéutico , Inhibidores de la Enzima Convertidora de Angiotensina/uso terapéutico , Espirometría
6.
Int J Mol Sci ; 24(6)2023 Mar 10.
Artículo en Inglés | MEDLINE | ID: mdl-36982382

RESUMEN

The pathophysiology of atrial fibrillation (AF) may involve atrial fibrosis/remodeling and dysfunctional endothelial activities. Despite the currently available treatment approaches, the progression of AF, its recurrence rate, and the high mortality risk of related complications underlay the need for more advanced prognostic and therapeutic strategies. There is increasing attention on the molecular mechanisms controlling AF onset and progression points to the complex cell to cell interplay that triggers fibroblasts, immune cells and myofibroblasts, enhancing atrial fibrosis. In this scenario, endothelial cell dysfunction (ED) might play an unexpected but significant role. microRNAs (miRNAs) regulate gene expression at the post-transcriptional level. In the cardiovascular compartment, both free circulating and exosomal miRNAs entail the control of plaque formation, lipid metabolism, inflammation and angiogenesis, cardiomyocyte growth and contractility, and even the maintenance of cardiac rhythm. Abnormal miRNAs levels may indicate the activation state of circulating cells, and thus represent a specific read-out of cardiac tissue changes. Although several unresolved questions still limit their clinical use, the ease of accessibility in biofluids and their prognostic and diagnostic properties make them novel and attractive biomarker candidates in AF. This article summarizes the most recent features of AF associated with miRNAs and relates them to potentially underlying mechanisms.


Asunto(s)
Fibrilación Atrial , Remodelación Atrial , MicroARNs , Enfermedades Vasculares , Humanos , MicroARNs/metabolismo , Fibrilación Atrial/genética , Fibrilación Atrial/complicaciones , Atrios Cardíacos/metabolismo , Biomarcadores/metabolismo , Enfermedades Vasculares/complicaciones , Fibrosis
7.
Medicina (Kaunas) ; 59(2)2023 Jan 19.
Artículo en Inglés | MEDLINE | ID: mdl-36837405

RESUMEN

Background and Objectives: COVID-19 induces massive systemic inflammation. Researchers have spent much time and effort finding an excellent and rapid image tool to evaluate COVID-19 patients. Since the pandemic's beginning, lung ultrasound (LUS) has been identified for this purpose. Monoclonal antibodies (mAb) were used to treat mild patients and prevent respiratory disease worsening. Materials and Methods: We evaluated 15 Caucasian patients with mild COVID-19 who did not require home oxygen, treated with Bamlanivimab and Etesevimab (Group 1). A molecular nose-throat swab test confirmed the diagnosis. All were office patients, and nobody was affected by respiratory failure. They were admitted to receive the single-day infusion of mAb treatment in agreement with the Italian Drug Agency (AIFA) rules for approval. LUS was performed before the drug administration (T0) and after three months (T1). We compared LUS at T1 in other outpatients who came for follow-up and were overlapping at the time of diagnosis for admittance criteria to receive mAb (Group 2). Results: Our COVID-19 outpatients reported no hospitalization in a follow-up visit after recovery. All patients became SARS-CoV-2 negative within one month since T0. LUS score at T0 was 8.23 ± 6.46. At T1 we found a significant decrease in Group 1 LUS score (5.18 ± 4.74; p < 0.05). We also found a significant decrease in the LUS score of Group 1 T1 compared to Group2 T1 (5.18 ± 4.74 vs 7.82 ± 5.21; p < 0.05). Conclusion: Early treatment of the SARS-CoV-2 virus effectively achieves a better recovery from disease and reduces lung involvement after three months as evaluated with LUS. Despite extrapolation to the general population may be done with caution, based on our data this ultrasound method is also effective for evaluating and following lung involvement in COVID-19 patients.


Asunto(s)
COVID-19 , Humanos , Proyectos Piloto , SARS-CoV-2 , Pulmón/diagnóstico por imagen , Ultrasonografía/métodos
8.
J Clin Immunol ; 42(5): 935-946, 2022 07.
Artículo en Inglés | MEDLINE | ID: mdl-35445287

RESUMEN

COVID-19 manifestations range from asymptomatic to life-threatening infections. The outcome in different inborn errors of immunity (IEI) is still a matter of debate. In this retrospective study, we describe the experience of the of the Italian Primary Immunodeficiencies Network (IPINet). Sixteen reference centers for adult or pediatric IEI were involved. One hundred fourteen patients were enrolled including 35 pediatric and 79 adult patients. Median age was 32 years, and male-to-female ratio was 1.5:1. The most common IEI were 22q11.2 deletion syndrome in children (26%) and common variable immunodeficiency (CVID) in adults (65%). Ninety-one patients did not require hospital admission, and among these, 33 were asymptomatic. Hospitalization rate was 20.17%. Older age (p 0.004) and chronic lung disease (p 0.0008) represented risk factors for hospitalization. Hospitalized patients mainly included adults suffering from humoral immunodeficiencies requiring immunoglobulin replacement therapy and as expected had lower B cell counts compared to non-hospitalized patients. Infection fatality rate in the whole cohort was 3.5%. Seroconversion was observed is 86.6% of the patients evaluated and in 83.3% of CVID patients. 16.85% of the patients reported long-lasting COVID symptoms. All but one patient with prolonged symptoms were under IgRT. The fatality rate observed in IEI was slightly similar to the general population. The age of the patients who did not survive was lower compared to the general population, and the age stratified mortality in the 50-60 age range considerable exceeded the mortality from 50 to 60 age group of the Italian population (14.3 vs 0.6%; p < 0.0001). We hypothesize that this is due to the fact that comorbidities in IEI patients are very common and usually appear early in life.


Asunto(s)
COVID-19 , Inmunodeficiencia Variable Común , Adulto , COVID-19/complicaciones , COVID-19/epidemiología , Niño , Inmunodeficiencia Variable Común/epidemiología , Femenino , Hospitalización , Humanos , Masculino , Estudios Retrospectivos , SARS-CoV-2 , Síndrome Post Agudo de COVID-19
9.
Am J Pathol ; 191(4): 748-758, 2021 04.
Artículo en Inglés | MEDLINE | ID: mdl-33516787

RESUMEN

Multiple myeloma (MM) progression closely depends on bone marrow (BM) angiogenesis. Several factors sustain angiogenesis, including cytokines, growth factors, and cell-to-cell interactions. Herein, BM thrombopoietin (TPO) was shown to support angiogenesis and disease progression in MM. Patients with MM at different progression phases had higher levels of BM and circulating TPO than monoclonal gammopathy of undetermined significance/smoldering MM patients, suggesting that TPO correlates with disease progression and prognosis. Endothelial cells from patients with monoclonal gammopathy of undetermined significance (MGECs) and endothelial cells from MM (MMECs) expressed TPO receptor, and the TPO treatment triggered their angiogenic capabilities in vitro. Indeed, TPO-treated MGECs and MMECs showed enhanced angiogenesis on Matrigel and spontaneous cell migration and chemotaxis by acting as a chemotactic agent. TPO also had an angiogenic activity in vivo in the chorioallantoic membrane assay system. Finally, TPO treatment increased the release of active matrix metalloproteinase (MMP)-9 and MMP-2 in MGECs and of MMP-2 in MMECs and affected the balance between angiogenic/antiangiogenic factors in the MM BM. Our results support the angiogenic activity of TPO, and suggest that it may have a critical role in promoting the angiogenic switch during MM progression. Accordingly, TPO may be envisaged as a new angiogenic and prognostic factor in patients with MM.


Asunto(s)
Células de la Médula Ósea/efectos de los fármacos , Progresión de la Enfermedad , Mieloma Múltiple/tratamiento farmacológico , Neovascularización Patológica/tratamiento farmacológico , Trombopoyetina/farmacología , Adulto , Anciano , Anciano de 80 o más Años , Inhibidores de la Angiogénesis/farmacología , Médula Ósea/efectos de los fármacos , Médula Ósea/metabolismo , Células de la Médula Ósea/metabolismo , Movimiento Celular/efectos de los fármacos , Células Endoteliales/efectos de los fármacos , Células Endoteliales/metabolismo , Femenino , Humanos , Masculino , Persona de Mediana Edad , Mieloma Múltiple/metabolismo , Neovascularización Patológica/metabolismo
10.
Nutr Metab Cardiovasc Dis ; 32(2): 420-428, 2022 02.
Artículo en Inglés | MEDLINE | ID: mdl-34893418

RESUMEN

BACKGROUND AND AIM: Left heart remodeling is a well-known pathophysiological effect of arterial hypertension. Right Heart status is not considered in its evaluation. No data are available on right atrium (RA) and its impact on the outcome in hypertension. We wondering to understand whether RA may play a role as a marker of an increased risk for organ damage in well-controlled hypertensives, to probe the clinical significance and whether it could indicate an increased risk. METHODS AND RESULTS: We studied well-controlled hypertensive patients. Heart damage was assessed by echocardiography. Patients were subdivided into those with RA area ≤18 cm2 (normal RA - Group 1) (554 pts, 227 M, aged 60.35 ± 10.48 years) and those >18 cm2 (Increased RA - Group 2) (101 pts, 71 M, age 61.65 ± 9.46 years). Group 2 had a higher left ventricle mass (LVM) and left atrium volume (LAV) both as absolute value (both p < 0.0001) and indexed for body surface area (LVMi p < 0.013; LAVi p = 0.0013). Group 2 showed an increased vascular stiffness (p < 0.0001) and carotid stenosis percentage (p = 0.011). TAPSE (p < 0.0001) resulted significantly increased. In The RA area was significantly correlated directly to LVM and LAV in both groups, but these correlations persisted in indexed values only in Group 2. Moreover, in this group there was a significant direct correlation between RA area and Tricuspid s'wave at echocardiography TDI analysis. Finally, Group 2 had an increased mortality rate compared to Group 1 (Log-Rank p = 0.0006). CONCLUSION: Group 2 hypertensive patients showed more alterations in dimensional and volumetric left heart parameters, and an increased mortality.


Asunto(s)
Lesiones Cardíacas , Hipertensión , Anciano , Cardiomegalia/diagnóstico por imagen , Ecocardiografía , Atrios Cardíacos/diagnóstico por imagen , Humanos , Hipertensión/complicaciones , Hipertensión/diagnóstico , Persona de Mediana Edad
11.
Int J Mol Sci ; 23(13)2022 Jun 30.
Artículo en Inglés | MEDLINE | ID: mdl-35806308

RESUMEN

Ion channels are pore-forming proteins that allow ions to flow across plasma membranes and intracellular organelles in both excitable and non-excitable cells. They are involved in the regulation of several biological processes (i.e., proliferation, cell volume and shape, differentiation, migration, and apoptosis). Recently, the aberrant expression of ion channels has emerged as an important step of malignant transformation, tumor progression, and drug resistance, leading to the idea of "onco-channelopathy". Here, we review the contribution of ion channels and transporters in multiple myeloma (MM), a hematological neoplasia characterized by the expansion of tumor plasma cells (MM cells) in the bone marrow (BM). Deregulation of ion channels sustains MM progression by modulating intracellular pathways that promote MM cells' survival, proliferation, and drug resistance. Finally, we focus on the promising role of ion channels as therapeutic targets for the treatment of MM patients in a combination strategy with currently used anti-MM drugs to improve their cytotoxic activity and reduce adverse effects.


Asunto(s)
Antineoplásicos , Mieloma Múltiple , Antineoplásicos/farmacología , Antineoplásicos/uso terapéutico , Apoptosis , Humanos , Canales Iónicos/metabolismo , Iones/metabolismo , Mieloma Múltiple/tratamiento farmacológico , Mieloma Múltiple/metabolismo
13.
Haematologica ; 106(7): 1943-1956, 2021 07 01.
Artículo en Inglés | MEDLINE | ID: mdl-32354870

RESUMEN

Interactions of malignant multiple myeloma (MM) plasma cells (MM-cells) with the microenvironment control MM-cell growth, survival, drug-resistance and dissemination. As in MM microvascular density increases in the bone marrow (BM), we investigated whether BM MM endothelial cells (MMECs) control disease progression via the junctional adhesion molecule A (JAM-A). Membrane and cytoplasmic JAM-A levels were upregulated in MMECs in 111 newly diagnosed (NDMM) and 201 relapsed-refractory (RRMM) patients compared to monoclonal gammopathy of undetermined significance (MGUS) and healthy controls. Elevated membrane expression of JAM-A on MMECs predicted poor clinical outcome. Mechanistically, addition of recombinant JAM-A to MMECs increased angiogenesis whereas its inhibition impaired angiogenesis and MM growth in 2D and 3D in vitro cell culture and chorioallantoic membrane-assays. To corroborate these findings, we treated MM bearing mice with JAM-A blocking mAb and demonstrated impaired MM progression corresponding to decreased MM-related vascularity. These findings support JAM-A as an important mediator of MM progression through facilitating MM-associated angiogenesis. Collectively, elevated JAM-A expression on bone marrow endothelial cells is an independent prognostic factor for patient survival in both NDMM and RRMM. Blocking JAM-A restricts angiogenesis in vitro, in embrio and in vivo and represents a suitable druggable molecule to halt neoangiogenesis and MM progression.


Asunto(s)
Molécula A de Adhesión de Unión , Mieloma Múltiple , Animales , Médula Ósea , Ecosistema , Células Endoteliales , Homeostasis , Humanos , Ratones , Mieloma Múltiple/tratamiento farmacológico , Microambiente Tumoral
14.
J Allergy Clin Immunol ; 146(2): 429-437, 2020 08.
Artículo en Inglés | MEDLINE | ID: mdl-32169379

RESUMEN

BACKGROUND: X-linked agammaglobulinemia (XLA) is the prototype of primary humoral immunodeficiencies. Long-term follow-up studies regarding disease-related complications and outcome are scarce. OBJECTIVE: Our aim was to describe the natural history of XLA. METHODS: A nationwide multicenter study based on the Italian Primary Immunodeficiency Network registry was established in 2000 in Italy. Affected patients were enrolled by documenting centers, and the patients' laboratory, clinical, and imaging data were recorded on an annual base. RESULTS: Data on the patients (N = 168) were derived from a cumulative follow-up of 1370 patient-years, with a mean follow-up of 8.35 years per patient. The mean age at diagnosis decreased after establishment of the Italian Primary Immunodeficiency Network registry (84 months before vs 23 months after). Respiratory, skin, and gastrointestinal manifestations were the most frequent clinical symptoms at diagnosis and during long-term follow-up. Regular immunoglobulin replacement treatment reduced the incidence of invasive infections. Affected patients developed chronic lung disease over time (47% after 40 years of follow-up) in the presence of chronic sinusitis (84%). Malignancies were documented in a minority of cases (3.7%). Overall survival for affected patients was significantly reduced when compared with that for the healthy male Italian population, and it further deteriorated in the presence of chronic lung disease. CONCLUSIONS: This is the first detailed long-term follow-up study for patients with XLA, revealing that although immunoglobulin replacement treatment reduces the incidence of invasive infections, it does not appear to influence the development of chronic lung disease. The overall survival of affected patients is reduced. Further studies are warranted to improve patients' clinical management and increase awareness among physicians.


Asunto(s)
Agammaglobulinemia/epidemiología , Enfermedades Genéticas Ligadas al Cromosoma X/epidemiología , Infecciones/epidemiología , Enfermedades Pulmonares/epidemiología , Sinusitis/epidemiología , Adolescente , Adulto , Agammaglobulinemia/mortalidad , Niño , Preescolar , Estudios de Seguimiento , Enfermedades Genéticas Ligadas al Cromosoma X/mortalidad , Humanos , Lactante , Recién Nacido , Italia/epidemiología , Masculino , Persona de Mediana Edad , Análisis de Supervivencia , Adulto Joven
15.
J Clin Immunol ; 40(7): 1026-1037, 2020 10.
Artículo en Inglés | MEDLINE | ID: mdl-32803625

RESUMEN

Primary immunodeficiencies (PIDs) are heterogeneous disorders, characterized by variable clinical and immunological features. National PID registries offer useful insights on the epidemiology, diagnosis, and natural history of these disorders. In 1999, the Italian network for primary immunodeficiencies (IPINet) was established. We report on data collected from the IPINet registry after 20 years of activity. A total of 3352 pediatric and adult patients affected with PIDs are registered in the database. In Italy, a regional distribution trend of PID diagnosis was observed. Based on the updated IUIS classification of 2019, PID distribution in Italy showed that predominantly antibody deficiencies account for the majority of cases (63%), followed by combined immunodeficiencies with associated or syndromic features (22.5%). The overall age at diagnosis was younger for male patients. The minimal prevalence of PIDs in Italy resulted in 5.1 per 100.000 habitants. Mortality was similar to other European registries (4.2%). Immunoglobulin replacement treatment was prescribed to less than one third of the patient cohort. Collectively, this is the first comprehensive description of the PID epidemiology in Italy.


Asunto(s)
Enfermedades de Inmunodeficiencia Primaria/epidemiología , Adolescente , Adulto , Niño , Preescolar , Bases de Datos Factuales , Femenino , Geografía Médica , Historia del Siglo XX , Historia del Siglo XXI , Humanos , Lactante , Recién Nacido , Italia/epidemiología , Masculino , Vigilancia de la Población , Prevalencia , Enfermedades de Inmunodeficiencia Primaria/diagnóstico , Enfermedades de Inmunodeficiencia Primaria/historia , Enfermedades de Inmunodeficiencia Primaria/terapia , Pronóstico , Sistema de Registros , Adulto Joven
16.
J Pathol ; 247(2): 241-253, 2019 02.
Artículo en Inglés | MEDLINE | ID: mdl-30357841

RESUMEN

Aberrant microRNA (miR) expression has an important role in tumour progression, but its involvement in bone marrow fibroblasts of multiple myeloma patients remains undefined. We demonstrate that a specific miR profile in bone marrow fibroblasts parallels the transition from monoclonal gammopathy of undetermined significance (MGUS) to myeloma. Overexpression of miR-27b-3p and miR-214-3p triggers proliferation and apoptosis resistance in myeloma fibroblasts via the FBXW7 and PTEN/AKT/GSK3 pathways, respectively. Transient transfection of miR-27b-3p and miR-214-3p inhibitors demonstrates a cooperation between these two miRNAs in the expression of the anti-apoptotic factor MCL1, suggesting that miR-27b-3p and miR-214-3p negatively regulate myeloma fibroblast apoptosis. Furthermore, myeloma cells modulate miR-27b-3p and miR-214-3p expression in fibroblasts through the release of exosomes. Indeed, tumour cell-derived exosomes induce an overexpression of both miRNAs in MGUS fibroblasts not through a simple transfer mechanism but by de novo synthesis triggered by the transfer of exosomal WWC2 protein that regulates the Hippo pathway. Increased levels of miR-27b-3p and miR-214-3p in MGUS fibroblasts co-cultured with myeloma cell-derived exosomes enhance the expression of fibroblast activation markers αSMA and FAP. These data show that the MGUS-to-myeloma transition entails an aberrant miRNA profile in marrow fibroblasts and highlight a key role of myeloma cells in modifying the bone marrow microenvironment by reprogramming the marrow fibroblasts' behaviour. Copyright © 2018 Pathological Society of Great Britain and Ireland. Published by John Wiley & Sons, Ltd.


Asunto(s)
Células de la Médula Ósea/metabolismo , Exosomas/metabolismo , Fibroblastos/metabolismo , MicroARNs/metabolismo , Gammopatía Monoclonal de Relevancia Indeterminada/metabolismo , Mieloma Múltiple/metabolismo , Actinas/metabolismo , Adulto , Anciano , Anciano de 80 o más Años , Apoptosis , Células de la Médula Ósea/patología , Células Cultivadas , Progresión de la Enfermedad , Endopeptidasas , Exosomas/genética , Exosomas/patología , Proteína 7 que Contiene Repeticiones F-Box-WD/genética , Proteína 7 que Contiene Repeticiones F-Box-WD/metabolismo , Femenino , Fibroblastos/patología , Gelatinasas/metabolismo , Regulación Neoplásica de la Expresión Génica , Humanos , Masculino , Proteínas de la Membrana/metabolismo , MicroARNs/genética , Persona de Mediana Edad , Gammopatía Monoclonal de Relevancia Indeterminada/genética , Gammopatía Monoclonal de Relevancia Indeterminada/patología , Mieloma Múltiple/genética , Mieloma Múltiple/patología , Fosfohidrolasa PTEN/genética , Fosfohidrolasa PTEN/metabolismo , Serina Endopeptidasas/metabolismo , Transducción de Señal , Microambiente Tumoral , Regulación hacia Arriba
17.
Mediators Inflamm ; 2020: 7527953, 2020.
Artículo en Inglés | MEDLINE | ID: mdl-32724296

RESUMEN

COVID-19 is a pandemic disease caused by the new coronavirus SARS-CoV-2 that mostly affects the respiratory system. The consequent inflammation is not able to clear viruses. The persistent excessive inflammatory response can build up a clinical picture that is very difficult to manage and potentially fatal. Modulating the immune response plays a key role in fighting the disease. One of the main defence systems is the activation of neutrophils that release neutrophil extracellular traps (NETs) under the stimulus of autophagy. Various molecules can induce NETosis and autophagy; some potent activators are damage-associated molecular patterns (DAMPs) and, in particular, the high-mobility group box 1 (HMGB1). This molecule is released by damaged lung cells and can induce a robust innate immunity response. The increase in HMGB1 and NETosis could lead to sustained inflammation due to SARS-CoV-2 infection. Therefore, blocking these molecules might be useful in COVID-19 treatment and should be further studied in the context of targeted therapy.


Asunto(s)
Alarminas/inmunología , Betacoronavirus , Infecciones por Coronavirus/tratamiento farmacológico , Infecciones por Coronavirus/inmunología , Trampas Extracelulares/inmunología , Neumonía Viral/tratamiento farmacológico , Neumonía Viral/inmunología , Alarminas/antagonistas & inhibidores , Autofagia/inmunología , Betacoronavirus/inmunología , COVID-19 , Infecciones por Coronavirus/patología , Trampas Extracelulares/efectos de los fármacos , Proteína HMGB1/antagonistas & inhibidores , Proteína HMGB1/inmunología , Interacciones Microbiota-Huesped/inmunología , Humanos , Interleucina-6/antagonistas & inhibidores , Interleucina-6/inmunología , Pulmón/inmunología , Pulmón/patología , Pandemias , Neumonía Viral/patología , SARS-CoV-2 , Tratamiento Farmacológico de COVID-19
18.
Int J Mol Sci ; 21(2)2020 Jan 17.
Artículo en Inglés | MEDLINE | ID: mdl-31963513

RESUMEN

Multiple myeloma is a B-cell lineage cancer in which neoplastic plasma cells expand in the bone marrow and pathophysiological interactions with components of microenvironment influence many biological aspects of the malignant phenotype, including apoptosis, survival, proliferation, and invasion. Despite the therapeutic progress achieved in the last two decades with the introduction of a more effective and safe new class of drugs (i.e., immunomodulators, proteasome inhibitors, monoclonal antibodies), there is improvement in patient survival, and multiple myeloma (MM) remains a non-curable disease. The bone marrow microenvironment is a complex structure composed of cells, extracellular matrix (ECM) proteins, and cytokines, in which tumor plasma cells home and expand. The role of the bone marrow (BM) microenvironment is fundamental during MM disease progression because modification induced by tumor plasma cells is crucial for composing a "permissive" environment that supports MM plasma cells proliferation, migration, survival, and drug resistance. The "activated phenotype" of the microenvironment of multiple myeloma is functional to plasma cell proliferation and spreading and to plasma cell drug resistance. Plasma cell drug resistance induced by bone marrow stromal cells is mediated by stress-managing pathways, autophagy, transcriptional rewiring, and non-coding RNAs dysregulation. These processes represent novel targets for the ever-increasing anti-MM therapeutic armamentarium.


Asunto(s)
Antineoplásicos/farmacología , Resistencia a Antineoplásicos , Células Madre Mesenquimatosas/patología , Mieloma Múltiple/tratamiento farmacológico , Mieloma Múltiple/patología , Animales , Humanos , Células Madre Mesenquimatosas/efectos de los fármacos , Microambiente Tumoral
19.
Int J Mol Sci ; 21(9)2020 Apr 26.
Artículo en Inglés | MEDLINE | ID: mdl-32357515

RESUMEN

: Immune checkpoint inhibitor (ICI)-related inflammatory diseases, including polymyositis (PM) and dermatomyositis (DM), in patients suffering from neoplastic disorders represent a medical challenge. The treatment of these conditions has taken on new urgency due to the successful and broad development of cancer-directed immunological-based therapeutic strategies. While primary and secondary PM/DM phenotypes have been pathophysiologically characterized, a rational, stepwise approach to the treatment of patients with ICI-related disease is lacking. In the absence of high-quality evidence to guide clinical judgment, the available data must be critically assessed. In this literature review, we examine partially neglected immunological and clinical findings to obtain insights into the biological profiles of ICI-related PM/DM and potential treatment options. We show that differential diagnosis is essential to stratifying patients according to prognosis and therapeutic impact. Finally, we provide a comprehensive assessment of druggable targets and suggest a stepwise patient-oriented approach for the treatment of ICI-related PM/DM.


Asunto(s)
Inhibidores de Puntos de Control Inmunológico/efectos adversos , Miositis/diagnóstico , Neoplasias/tratamiento farmacológico , Diagnóstico Diferencial , Manejo de la Enfermedad , Humanos , Inhibidores de Puntos de Control Inmunológico/uso terapéutico , Miositis/inducido químicamente , Miositis/inmunología , Fenotipo
20.
Int J Mol Sci ; 21(9)2020 Apr 27.
Artículo en Inglés | MEDLINE | ID: mdl-32349317

RESUMEN

MicroRNAs (miRNAs, or miRs) are single-strand short non-coding RNAs with a pivotal role in the regulation of physiological- or disease-associated cellular processes. They bind to target miRs modulating gene expression at post-transcriptional levels. Here, we present an overview of miRs deregulation in the pathogenesis of multiple myeloma (MM), and discuss the potential use of miRs/nanocarriers association in clinic. Since miRs can act as oncogenes or tumor suppressors, strategies based on their inhibition and/or replacement represent the new opportunities in cancer therapy. The miRs delivery systems include liposomes, polymers, and exosomes that increase their physical stability and prevent nuclease degradation. Phase I/II clinical trials support the importance of miRs as an innovative therapeutic approach in nanomedicine to prevent cancer progression and drug resistance. Results in clinical practice are promising.


Asunto(s)
Resistencia a Antineoplásicos/genética , Técnicas de Transferencia de Gen , MicroARNs/genética , Mieloma Múltiple/genética , Mieloma Múltiple/terapia , Nanotecnología , Tratamiento con ARN de Interferencia , Animales , Progresión de la Enfermedad , Exosomas , Regulación Neoplásica de la Expresión Génica , Humanos , Lípidos/química , Liposomas , Mieloma Múltiple/patología , Nanotecnología/métodos , Polímeros/química , Tratamiento con ARN de Interferencia/métodos
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA