Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 792
Filtrar
1.
Handb Clin Neurol ; 202: 23-39, 2024.
Artículo en Inglés | MEDLINE | ID: mdl-39111909

RESUMEN

Hematopoiesis is a complex process that takes place inside the bone marrow, where a specialized structure, the bone marrow niche, participates in the maintenance of hematopoietic stem cell functionality. Inflammatory conditions, such as autoimmune diseases, could alter this equilibrium leading to pathologic consequences. Immune cells, which also reside in the bone marrow, directly participate in sustaining the inflammatory state in autoimmune diseases. In particular, memory lymphocytes are key players in the long-term maintenance of the immune response against self-antigens, causing tissue damage and bone marrow alterations.


Asunto(s)
Enfermedades Autoinmunes , Humanos , Enfermedades Autoinmunes/inmunología , Animales , Memoria Inmunológica/inmunología , Hematopoyesis/fisiología , Hematopoyesis/inmunología , Células Madre Hematopoyéticas/inmunología
2.
Trends Immunol ; 45(8): 597-608, 2024 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-39030115

RESUMEN

Innate immune cells are primary effectors during host defense and in sterile inflammation. Their production in the bone marrow is tightly regulated by growth and niche factors, and their activity at sites of inflammation is orchestrated by a network of alarmins and cytokines. Yet, recent work highlights a significant role of the peripheral nervous system in these processes. Sympathetic neural pathways play a key role in regulating blood cell homeostasis, and sensory neural pathways mediate pro- or anti-inflammatory signaling in a tissue-specific manner. Here, we review emerging evidence of the fine titration of hematopoiesis, leukocyte trafficking, and tissue repair via neuro-immune crosstalk, and how its derailment can accelerate chronic inflammation, as in atherosclerosis.


Asunto(s)
Hematopoyesis , Inflamación , Neuroinmunomodulación , Humanos , Inflamación/inmunología , Animales , Hematopoyesis/inmunología , Inmunidad Innata , Transducción de Señal/inmunología
3.
Eur J Immunol ; 54(8): e2350851, 2024 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-38803021

RESUMEN

Stress exposure has been shown to modulate innate and adaptive immune responses. Indeed, stress favors myelopoiesis and monocyte generation and contributes to cardiovascular disease development. As sex hormones regulate innate and adaptive immune responses, we decided to investigate whether stress exposure leads to a different immune response in female and male mice. Our data demonstrated that psychosocial stressinduced neutrophilia in male, but not female mice. Importantly, we identified that B-cell numbers were reduced in female, but not male mice upon exposure to stress. Thus, our study revealed that the stress-induced immune alterations are sex-dependent, and this is an important feature to consider for future investigations.


Asunto(s)
Hematopoyesis , Estrés Psicológico , Animales , Femenino , Estrés Psicológico/inmunología , Masculino , Ratones , Hematopoyesis/inmunología , Linfocitos B/inmunología , Neutrófilos/inmunología , Leucocitos/inmunología , Ratones Endogámicos C57BL , Factores Sexuales , Caracteres Sexuales
4.
J Mol Biol ; 435(15): 168169, 2023 08 01.
Artículo en Inglés | MEDLINE | ID: mdl-37263392

RESUMEN

Bacille Calmette-Guérin (BCG) is the most commonly administered vaccine in human history. The medical application of BCG extends far beyond the fight against tuberculosis. Despite its stellar medical record over 100 years, insight into how BCG provides this vast range of benefits is largely limited, both for its pathogen-specific (tuberculosis) as well as pathogen-agnostic (other infections, autoimmunity, allergies, and cancer) effects. Trained immunity and emergency granulopoiesis have been identified as mediating BCG's pathogen-agnostic effects, for which some of the molecular mechanisms have been delineated. Upon review of the existing evidence, we postulate that emergency granulopoiesis and trained immunity are a continuum of the same effect cascade. In this context, we highlight that BCG's pathogen-agnostic benefits could be optimized by taking advantage of the age of the recipient and route of BCG administration.


Asunto(s)
Vacuna BCG , Hematopoyesis , Inmunidad Entrenada , Tuberculosis , Humanos , Vacuna BCG/inmunología , Hematopoyesis/inmunología , Tuberculosis/prevención & control
5.
J Virol ; 97(4): e0010223, 2023 04 27.
Artículo en Inglés | MEDLINE | ID: mdl-37022164

RESUMEN

Whether and how a local virus infection affects the hematopoietic system in the bone marrow is largely unknown, unlike with systemic infection. In this study, we showed that influenza A virus (IAV) infection leads to demand-adapted monopoiesis in the bone marrow. The beta interferon (IFN-ß) promoter stimulator 1 (IPS-1)-type I IFN-IFN-α receptor 1 (IFNAR1) axis-mediated signaling was found to induce the emergency expansion of the granulocyte-monocyte progenitor (GMP) population and upregulate the expression of the macrophage colony-stimulating factor receptor (M-CSFR) on bipotent GMPs and monocyte progenitors via the signal transducer and activator of transcription 1 (STAT1), leading to a scaled-back proportion of granulocyte progenitors. To further address the influence of demand-adapted monopoiesis on IAV-induced secondary bacterial infection, IAV-infected wild-type (WT) and Stat1-/- mice were challenged with Streptococcus pneumoniae. Compared with WT mice, Stat1-/- mice did not demonstrate demand-adapted monopoiesis, had more infiltrating granulocytes, and were able to effectively eliminate the bacterial infection. IMPORTANCE Our findings show that influenza A virus infection induces type I interferon (IFN)-mediated emergency hematopoiesis to expand the GMP population in the bone marrow. The type I IFN-STAT1 axis was identified as being involved in mediating the viral-infection-driven demand-adapted monopoiesis by upregulating M-CSFR expression in the GMP population. As secondary bacterial infections often manifest during a viral infection and can lead to severe or even fatal clinical complications, we further assessed the impact of the observed monopoiesis on bacterial clearance. Our results suggest that the resulting decrease in the proportion of granulocytes may play a role in diminishing the IAV-infected host's ability to effectively clear secondary bacterial infection. Our findings not only provide a more complete picture of the modulatory functions of type I IFN but also highlight the need for a more comprehensive understanding of potential changes in hematopoiesis during local infections to better inform clinical interventions.


Asunto(s)
Interferón Tipo I , Infecciones por Orthomyxoviridae , Receptor de Factor Estimulante de Colonias de Macrófagos , Factor de Transcripción STAT1 , Regulación hacia Arriba , Animales , Humanos , Ratones , Virus de la Influenza A/inmunología , Interferón Tipo I/inmunología , Receptor de Factor Estimulante de Colonias de Macrófagos/genética , Receptor de Factor Estimulante de Colonias de Macrófagos/inmunología , Factor de Transcripción STAT1/genética , Factor de Transcripción STAT1/inmunología , Infecciones por Orthomyxoviridae/inmunología , Hematopoyesis/inmunología , Células Progenitoras de Granulocitos y Macrófagos/inmunología , Streptococcus pneumoniae/inmunología , Infecciones Neumocócicas/inmunología
6.
Scand J Immunol ; 95(3): e13130, 2022 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-34951041

RESUMEN

T cell receptor excision circles (TRECs) and kappa-deleting excision circles (KRECs) are DNA fragments potentially indicative of T and B cell development, respectively. Recent thymic emigrants (RTEs) are a subset of peripheral cells that may also represent thymic function. Here, we investigated TREC/KREC copy numbers by quantitative real-time PCR in the peripheral blood of patients with primary immunodeficiencies (PIDs, n = 145) and that of healthy controls (HCs, n = 86) and assessed the correlation between RTEs and TREC copy numbers. We found that TREC copy numbers were significantly lower in children and adults with PIDs (P < .0001 and P < .002, respectively) as compared with their respective age-matched HCs. A moderate correlation was observed between TREC copies and RTE numbers among children with PID (r = .5114, P < .01), whereas no significant correlation was detected between RTE values and TREC content in the HCs (r = .0205, P = .9208). Additionally, we determined TREC and KREC copy numbers in DNA isolated from the Guthrie cards of 200 newborns and showed that this method is applicable to DNA isolated from both peripheral blood samples and dried blood spots, with the two sample types showing comparable TREC and KREC values. We further showed that RTE values are not always reliable markers of T cell output. Although additional confirmatory studies with larger cohorts are needed, our results provide thresholds for TREC/KREC copy numbers for different age groups.


Asunto(s)
Linfocitos B/inmunología , Receptores de Antígenos de Linfocitos T/inmunología , Inmunodeficiencia Combinada Grave/inmunología , Linfocitos T/inmunología , Adolescente , Adulto , Niño , Preescolar , ADN/genética , ADN/inmunología , Femenino , Hematopoyesis/genética , Hematopoyesis/inmunología , Humanos , Lactante , Recién Nacido , Activación de Linfocitos/inmunología , Masculino , Persona de Mediana Edad , Tamizaje Neonatal/métodos , Reacción en Cadena en Tiempo Real de la Polimerasa/métodos , Receptores de Antígenos de Linfocitos T/genética , Inmunodeficiencia Combinada Grave/genética , Adulto Joven
7.
Toxicology ; 464: 152990, 2021 12.
Artículo en Inglés | MEDLINE | ID: mdl-34673135

RESUMEN

Benzene is a typical hematopoietic toxic substance, that can cause serious blood and circulatory system diseases such as aplastic anemia, myelodysplastic syndrome and acute myeloid leukemia, but the immunological mechanism by which this occurs is not clear. T helper cells play a key role in regulating the immune balance in the body. In this study, benzene-induced hematopoietic toxicity BALB/c mice model was established, and changes in immune organs and T helper cell subsets (Th1, Th2, Th17 and Treg cells) were explored. At 28 days after subcutaneous injection of 150 mg/kg benzene, mice showed pancytopenia and obvious pathological damage to the bone marrow, spleen, and thymus. Flow cytometry revealed that the number of CD4+CD25+Foxp3+ Treg cells in the spleen increased significantly. The level of IL-10 in the spleen, serum, and bone marrow increased, while the levels of IL-17 in the spleen and serum decreased. Furthermore, the levels of CD4 and CD8 proteins in the spleen decreased. Immunofluorescence results showed that levels of Foxp3, a specific transcription factor that induced the differentiation of Treg cells, increased after exposure to benzene. Our results demonstrate that immunosuppression occurred in the benzene-induced hematopoietic toxicity model mice, and Treg cells and secreted IL-10 may play a key role in the process.


Asunto(s)
Benceno/toxicidad , Hematopoyesis/efectos de los fármacos , Interleucina-10/inmunología , Linfocitos T Reguladores/inmunología , Animales , Médula Ósea/efectos de los fármacos , Médula Ósea/patología , Diferenciación Celular/efectos de los fármacos , Hematopoyesis/inmunología , Tolerancia Inmunológica/efectos de los fármacos , Masculino , Ratones , Ratones Endogámicos BALB C , Bazo/efectos de los fármacos , Bazo/patología , Linfocitos T Colaboradores-Inductores/inmunología , Timo/efectos de los fármacos , Timo/patología
8.
Front Immunol ; 12: 714244, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-34552585

RESUMEN

Platinum-based chemotherapy is an effective treatment used in multiple tumor treatments, but produces severe side effects including neurotoxicity, anemia, and immunosuppression, which limits its anti-tumor efficacy and increases the risk of infections. Electroacupuncture (EA) is often used to ameliorate these side effects, but its mechanism is unknown. Here, we report that EA on ST36 and SP6 prevents cisplatin-induced neurotoxicity and immunosuppression. EA induces neuroprotection, prevents pain-related neurotoxicity, preserves bone marrow (BM) hematopoiesis, and peripheral levels of leukocytes. EA activates sympathetic BM terminals to release pituitary adenylate cyclase activating polypeptide (PACAP). PACAP-receptor PAC1-antagonists abrogate the effects of EA, whereas PAC1-agonists mimic EA, prevent neurotoxicity, immunosuppression, and preserve BM hematopoiesis during cisplatin chemotherapy. Our results indicate that PAC1-agonists may provide therapeutic advantages during chemotherapy to treat patients with advanced neurotoxicity or neuropathies limiting EA efficacy.


Asunto(s)
Cisplatino/uso terapéutico , Electroacupuntura , Inmunomodulación , Neuroinmunomodulación , Receptores del Polipéptido Activador de la Adenilato-Ciclasa Hipofisaria/genética , Animales , Células de la Médula Ósea/metabolismo , Neutropenia Febril Inducida por Quimioterapia , Cisplatino/farmacología , Manejo de la Enfermedad , Modelos Animales de Enfermedad , Electroacupuntura/métodos , Hematopoyesis/genética , Hematopoyesis/inmunología , Humanos , Inmunomodulación/genética , Leucopenia , Ratones , Neoplasias/genética , Neoplasias/inmunología , Neoplasias/terapia , Neuroinmunomodulación/genética , Receptores del Polipéptido Activador de la Adenilato-Ciclasa Hipofisaria/metabolismo
9.
Sci Rep ; 11(1): 18046, 2021 09 10.
Artículo en Inglés | MEDLINE | ID: mdl-34508131

RESUMEN

Chronic myeloid leukemia (CML) is a form of myeloproliferative neoplasm caused by the oncogenic tyrosine kinase BCR-ABL. Although tyrosine kinase inhibitors have dramatically improved the prognosis of patients with CML, several problems such as resistance and recurrence still exist. Immunological control may contribute to solving these problems, and it is important to understand why CML patients fail to spontaneously develop anti-tumor immunity. Here, we show that differentiation of conventional dendritic cells (cDCs), which are vital for anti-tumor immunity, is restricted from an early stage of hematopoiesis in CML. In addition, we found that monocytes and basophils, which are increased in CML patients, express high levels of PD-L1, an immune checkpoint molecule that inhibits T cell responses. Moreover, RNA-sequencing analysis revealed that basophils express genes related to poor prognosis in CML. Our data suggest that BCR-ABL not only disrupts the "accelerator" (i.e., cDCs) but also applies the "brake" (i.e., monocytes and basophils) of anti-tumor immunity, compromising the defense against CML cells.


Asunto(s)
Leucemia Mielógena Crónica BCR-ABL Positiva/etiología , Leucemia Mielógena Crónica BCR-ABL Positiva/patología , Microambiente Tumoral/inmunología , Adulto , Anciano , Anciano de 80 o más Años , Animales , Biomarcadores , Médula Ósea/inmunología , Médula Ósea/metabolismo , Médula Ósea/patología , Carcinogénesis/genética , Carcinogénesis/inmunología , Biología Computacional/métodos , Bases de Datos Genéticas , Células Dendríticas/inmunología , Células Dendríticas/metabolismo , Células Dendríticas/patología , Modelos Animales de Enfermedad , Susceptibilidad a Enfermedades , Femenino , Perfilación de la Expresión Génica , Hematopoyesis/genética , Hematopoyesis/inmunología , Humanos , Inmunidad/genética , Inmunofenotipificación , Leucemia Mielógena Crónica BCR-ABL Positiva/mortalidad , Masculino , Ratones , Persona de Mediana Edad , Estadificación de Neoplasias , Neutrófilos/inmunología , Neutrófilos/metabolismo , Transcriptoma , Microambiente Tumoral/genética , Adulto Joven
10.
Cells ; 10(7)2021 06 22.
Artículo en Inglés | MEDLINE | ID: mdl-34206607

RESUMEN

The ubiquitin-proteasome system (UPS) is a central part of protein homeostasis, degrading not only misfolded or oxidized proteins but also proteins with essential functions. The fact that a healthy hematopoietic system relies on the regulation of protein homeostasis and that alterations in the UPS can lead to malignant transformation makes the UPS an attractive therapeutic target for the treatment of hematologic malignancies. Herein, inhibitors of the proteasome, the last and most important component of the UPS enzymatic cascade, have been approved for the treatment of these malignancies. However, their use has been associated with side effects, drug resistance, and relapse. Inhibitors of the immunoproteasome, a proteasomal variant constitutively expressed in the cells of hematopoietic origin, could potentially overcome the encountered problems of non-selective proteasome inhibition. Immunoproteasome inhibitors have demonstrated their efficacy and safety against inflammatory and autoimmune diseases, even though their development for the treatment of hematologic malignancies is still in the early phases. Various immunoproteasome inhibitors have shown promising preliminary results in pre-clinical studies, and one inhibitor is currently being investigated in clinical trials for the treatment of multiple myeloma. Here, we will review data on immunoproteasome function and inhibition in hematopoietic cells and hematologic cancers.


Asunto(s)
Neoplasias Hematológicas/enzimología , Neoplasias Hematológicas/inmunología , Hematopoyesis/inmunología , Complejo de la Endopetidasa Proteasomal/inmunología , Neoplasias Hematológicas/tratamiento farmacológico , Hematopoyesis/efectos de los fármacos , Humanos , Complejo de la Endopetidasa Proteasomal/genética , Inhibidores de Proteasoma/farmacología , Inhibidores de Proteasoma/uso terapéutico , Transducción de Señal/efectos de los fármacos
11.
J Trauma Acute Care Surg ; 91(4): 692-699, 2021 10 01.
Artículo en Inglés | MEDLINE | ID: mdl-34252063

RESUMEN

BACKGROUND: After severe trauma, the older host experiences more dysfunctional hematopoiesis of bone marrow (BM) hematopoietic stem and progenitor cells (HSPCs), and dysfunctional differentiation of circulating myeloid cells into effective innate immune cells. Our main objective was to compare BM HSPC microRNA (miR) responses of old and young mice in a clinically relevant model of severe trauma and shock. METHODS: C57BL/6 adult male mice aged 8 to 12 weeks (young) and 18 to 24 months (old) underwent multiple injuries and hemorrhagic shock (polytrauma [PT]) that engenders the equivalent of major trauma (Injury Severity Score, >15). Pseudomonas pneumonia (PNA) was induced in some young and old adult mice 24 hours after PT. MicroRNA expression patterns were determined from lineage-negative enriched BM HSPCs isolated from PT and PT-PNA mice at 24 and 48 hours postinjury, respectively. Genome-wide expression and pathway analyses were also performed on bronchoalveolar lavage (BAL) leukocytes from both mouse cohorts. RESULTS: MicroRNA expression significantly differed among all experimental conditions (p < 0.05), except for old-naive versus old-injured (PT or PT-PNA) mice, suggesting an inability of old mice to mount a robust early miR response to severe shock and injury. In addition, young adult mice had significantly more leukocytes obtained from their BAL, and there were greater numbers of polymorphonuclear cells compared with old mice (59.8% vs. 2.2%, p = 0.0069). Despite increased gene expression changes, BAL leukocytes from old mice demonstrated a more dysfunctional transcriptomic response to PT-PNA than young adult murine BAL leukocytes, as reflected in predicted upstream functional pathway analysis. CONCLUSION: The miR expression pattern in BM HSPCs after PT (+/-PNA) is dissimilar in old versus young adult mice. In the acute postinjury phase, old adult mice are unable to mount a robust miR HSPC response. Hematopoietic stem and progenitor cell miR expression in old PT mice reflects a diminished functional status and a blunted capacity for terminal differentiation of myeloid cells.


Asunto(s)
Médula Ósea/patología , Hematopoyesis/genética , Células Madre Hematopoyéticas/fisiología , Traumatismo Múltiple/complicaciones , Choque Hemorrágico/inmunología , Factores de Edad , Envejecimiento/sangre , Envejecimiento/genética , Envejecimiento/inmunología , Animales , Médula Ósea/fisiología , Diferenciación Celular/inmunología , Modelos Animales de Enfermedad , Perfilación de la Expresión Génica , Regulación de la Expresión Génica/inmunología , Hematopoyesis/inmunología , Humanos , Inmunidad Innata , Masculino , Ratones , Ratones Endogámicos C57BL , Traumatismo Múltiple/sangre , Traumatismo Múltiple/inmunología , Choque Hemorrágico/sangre , Choque Hemorrágico/genética , Choque Hemorrágico/patología
12.
J Cancer Res Ther ; 17(2): 547-550, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-34121706

RESUMEN

PURPOSE: Health emergency due to COVID-19 started in Uruguay on March 13, 2020; our mastology unit tried to ensure adequate oncological care, and protect patients from the virus infection and complications. OBJECTIVE: To assess the health care activities in the "peak" of the pandemic during 3 months. MATERIALS AND METHODS: we collected data from the electronic health record. RESULTS: There were a total of 293 medical appointments from 131 patients (221 face-to-face), that decreased by 16.7% compared to the same period in 2019 (352 appointments). The medical appointments were scheduled to evaluate the continuity of systemic treatment or modifications (95 patients; 72.5%), follow-up (17; 12.9%), first-time consultation (12; 9.1%), and assess paraclinical studies (7; 5.3%). The patients were on hormone therapy (81 patients; 74%), chemotherapy (CT) (21; 19%), and anti-HER2 therapies (9; 8%). New twenty treatments were initiated. Of the 14 patients that were on adjuvant/neoadjuvant CT, 9 (64.3%) continued with the same regimen with the addition of prophylactic granulocyte-colony-stimulating factors (G-CSF), and 5 (35.7%), who were receiving weekly paclitaxel, continued the treatment with no changes. Of the seven patients that were on palliative CT, 2 (28.5%) continued the treatment with the addition of G-CSF, 3 (42.8%) continued with weekly capecitabine or paclitaxel with no treatment changes, and 2 (28.5%) changed their treatment regimen (a less myelosuppressive regimen was selected for one and due to progression of the disease in the other patient). The ninety patients who were receiving adjuvant, neoadjuvant, or palliative criteria hormone therapy and/or anti-HER2 therapies, continued the treatment with no changes. CONCLUSIONS: The evidence suggests that, although medical appointments decreased by approximately 17%, we could maintain healthcare activities, continued most of the treatments while the most modified was CT with G-CSF to avoid myelosuppression.


Asunto(s)
Neoplasias de la Mama/tratamiento farmacológico , COVID-19/epidemiología , Continuidad de la Atención al Paciente/estadística & datos numéricos , Atención a la Salud/estadística & datos numéricos , Oncología Médica/estadística & datos numéricos , Adulto , Anciano , Anciano de 80 o más Años , Protocolos de Quimioterapia Combinada Antineoplásica/efectos adversos , Médula Ósea/efectos de los fármacos , Neoplasias de la Mama/complicaciones , Neoplasias de la Mama/diagnóstico , Neoplasias de la Mama/inmunología , COVID-19/inmunología , COVID-19/prevención & control , COVID-19/transmisión , Control de Enfermedades Transmisibles/normas , Continuidad de la Atención al Paciente/organización & administración , Atención a la Salud/organización & administración , Atención a la Salud/normas , Registros Electrónicos de Salud/estadística & datos numéricos , Femenino , Factor Estimulante de Colonias de Granulocitos/administración & dosificación , Hematopoyesis/efectos de los fármacos , Hematopoyesis/inmunología , Humanos , Oncología Médica/organización & administración , Oncología Médica/normas , Persona de Mediana Edad , Pandemias/prevención & control , Derivación y Consulta/normas , Derivación y Consulta/estadística & datos numéricos , Estudios Retrospectivos , Telemedicina/organización & administración , Telemedicina/normas , Telemedicina/estadística & datos numéricos , Triaje/organización & administración , Triaje/normas , Uruguay/epidemiología
13.
Sci Rep ; 11(1): 11452, 2021 06 01.
Artículo en Inglés | MEDLINE | ID: mdl-34075076

RESUMEN

Using a ground-based model to simulate spaceflight [21-days of single-housed, hindlimb unloading (HLU) combined with continuous low-dose gamma irradiation (LDR, total dose of 0.04 Gy)], an in-depth survey of the immune and hematological systems of mice at 7-days post-exposure was performed. Collected blood was profiled with a hematology analyzer and spleens were analyzed by whole transcriptome shotgun sequencing (RNA-sequencing). The results revealed negligible differences in immune differentials. However, hematological system analyses of whole blood indicated large disparities in red blood cell differentials and morphology, suggestive of anemia. Murine Reactome networks indicated majority of spleen cells displayed differentially expressed genes (DEG) involved in signal transduction, metabolism, cell cycle, chromatin organization, and DNA repair. Although immune differentials were not changed, DEG analysis of the spleen revealed expression profiles associated with inflammation and dysregulated immune function persist to 1-week post-simulated spaceflight. Additionally, specific regulation pathways associated with human blood disease gene orthologs, such as blood pressure regulation, transforming growth factor-ß receptor signaling, and B cell differentiation were noted. Collectively, this study revealed differential immune and hematological outcomes 1-week post-simulated spaceflight conditions, suggesting recovery from spaceflight is an unremitting process.


Asunto(s)
Rayos gamma/efectos adversos , Hematopoyesis/inmunología , Hematopoyesis/efectos de la radiación , Suspensión Trasera , Transducción de Señal/efectos de la radiación , Animales , Relación Dosis-Respuesta en la Radiación , Femenino , Ratones
14.
J Exp Med ; 218(7)2021 07 05.
Artículo en Inglés | MEDLINE | ID: mdl-34129017

RESUMEN

With a growing aged population, there is an imminent need to develop new therapeutic strategies to ameliorate disorders of hematopoietic aging, including clonal hematopoiesis and myelodysplastic syndrome (MDS). Cell-intrinsic dysregulation of innate immune- and inflammatory-related pathways as well as systemic inflammation have been implicated in hematopoietic defects associated with aging, clonal hematopoiesis, and MDS. Here, we review and discuss the role of dysregulated innate immune and inflammatory signaling that contribute to the competitive advantage and clonal dominance of preleukemic and MDS-derived hematopoietic cells. We also propose how emerging concepts will further reveal critical biology and novel therapeutic opportunities.


Asunto(s)
Envejecimiento/inmunología , Hematopoyesis/inmunología , Células Madre Hematopoyéticas/inmunología , Inmunidad Innata/inmunología , Inflamación/inmunología , Síndromes Mielodisplásicos/inmunología , Animales , Hematopoyesis Clonal/inmunología , Humanos , Transducción de Señal/inmunología
15.
Front Immunol ; 12: 661900, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-34054827

RESUMEN

TNF is a multifunctional cytokine with its key functions attributed to inflammation, secondary lymphoid tissue organogenesis and immune regulation. However, it is also a physiological regulator of hematopoiesis and is involved in development and homeostatic maintenance of various organs and tissues. Somewhat unexpectedly, the most important practical application of TNF biology in medicine is anti-TNF therapy in several autoimmune diseases. With increased number of patients undergoing treatment with TNF inhibitors and concerns regarding possible adverse effects of systemic cytokine blockade, the interest in using humanized mouse models to study the efficacy and safety of TNF-targeting biologics in vivo is justified. This Perspective discusses the main functions of TNF and its two receptors, TNFR1 and TNFR2, in steady state, as well as in emergency hematopoiesis. It also provides a comparative overview of existing mouse lines with humanization of TNF/TNFR system. These genetically engineered mice allow us to study TNF signaling cascades in the hematopoietic compartment in the context of various experimental disease models and for evaluating the effects of various human TNF inhibitors on hematopoiesis and other physiological processes.


Asunto(s)
Hematopoyesis/efectos de los fármacos , Hematopoyesis/inmunología , Factor de Necrosis Tumoral alfa/antagonistas & inhibidores , Factor de Necrosis Tumoral alfa/inmunología , Animales , Células Cultivadas , Humanos , Inflamación/inmunología , Ratones , Ratones Transgénicos , Receptores Tipo I de Factores de Necrosis Tumoral/antagonistas & inhibidores , Receptores Tipo II del Factor de Necrosis Tumoral/antagonistas & inhibidores , Transducción de Señal
16.
Cell Rep ; 35(2): 108967, 2021 04 13.
Artículo en Inglés | MEDLINE | ID: mdl-33852867

RESUMEN

T lymphocyte differentiation in the steady state is characterized by high cellular turnover whereby thymocytes do not self-renew. However, if deprived of competent progenitors, the thymus can temporarily maintain thymopoiesis autonomously. This bears a heavy cost, because prolongation of thymus autonomy causes leukemia. Here, we show that, at an early stage, thymus autonomy relies on double-negative 3 early (DN3e) thymocytes that acquire stem-cell-like properties. Following competent progenitor deprivation, DN3e thymocytes become long lived, are required for thymus autonomy, differentiate in vivo, and include DNA-label-retaining cells. At the single-cell level, the transcriptional programs of thymopoiesis in autonomy and the steady state are similar. However, a new cell population emerges in autonomy that expresses an aberrant Notch target gene signature and bypasses the ß-selection checkpoint. In summary, DN3e thymocytes have the potential to self-renew and differentiate in vivo if cell competition is impaired, but this generates atypical cells, probably the precursors of leukemia.


Asunto(s)
Hematopoyesis/genética , Leucemia/genética , Receptores Notch/genética , Timocitos/inmunología , Timo/inmunología , Factores de Transcripción/genética , Animales , Diferenciación Celular , Proliferación Celular , Familia de Proteínas EGF/genética , Familia de Proteínas EGF/inmunología , Perfilación de la Expresión Génica , Regulación de la Expresión Génica , Hematopoyesis/inmunología , Humanos , Inmunofenotipificación , Riñón , Leucemia/inmunología , Leucemia/patología , Activación de Linfocitos , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Receptores Notch/inmunología , Transducción de Señal , Análisis de la Célula Individual , Timocitos/clasificación , Timocitos/patología , Timo/patología , Timo/trasplante , Factores de Transcripción/inmunología , Trasplante Heterotópico , Trasplante Homólogo
17.
Mucosal Immunol ; 14(5): 1017-1037, 2021 09.
Artículo en Inglés | MEDLINE | ID: mdl-33859369

RESUMEN

The mechanisms that maintain intestinal homeostasis despite constant exposure of the gut surface to multiple environmental antigens and to billions of microbes have been scrutinized over the past 20 years with the goals to gain basic knowledge, but also to elucidate the pathogenesis of inflammatory bowel diseases (IBD) and to identify therapeutic targets for these severe diseases. Considerable insight has been obtained from studies based on gene inactivation in mice as well as from genome wide screens for genetic variants predisposing to human IBD. These studies are, however, not sufficient to delineate which pathways play key nonredundant role in the human intestinal barrier and to hierarchize their respective contribution. Here, we intend to illustrate how such insight can be derived from the study of human Mendelian diseases, in which severe intestinal pathology results from single gene defects that impair epithelial and or hematopoietic immune cell functions. We suggest that these diseases offer the unique opportunity to study in depth the pathogenic mechanisms leading to perturbation of intestinal homeostasis in humans. Furthermore, molecular dissection of monogenic intestinal diseases highlights key pathways that might be druggable and therapeutically targeted in common forms of IBD.


Asunto(s)
Interacciones Microbiota-Huesped/inmunología , Inmunidad Mucosa , Inmunomodulación , Mucosa Intestinal/fisiología , Inmunidad Adaptativa , Animales , Transporte Biológico , Biomarcadores , Diferenciación Celular/genética , Diferenciación Celular/inmunología , Susceptibilidad a Enfermedades , Enterocitos/citología , Enterocitos/inmunología , Enterocitos/metabolismo , Regulación de la Expresión Génica , Enfermedades Genéticas Congénitas/genética , Enfermedades Genéticas Congénitas/inmunología , Predisposición Genética a la Enfermedad , Hematopoyesis/genética , Hematopoyesis/inmunología , Homeostasis , Interacciones Microbiota-Huesped/genética , Humanos , Inmunidad Innata , Mutación , Transducción de Señal
18.
Front Immunol ; 12: 643852, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-33692812

RESUMEN

Since the late 1980s, mice have been repopulated with human hematopoietic cells to study the fundamental biology of human hematopoiesis and immunity, as well as a broad range of human diseases in vivo. Multiple mouse recipient strains have been developed and protocols optimized to efficiently generate these "humanized" mice. Here, we review three guiding principles that have been applied to the development of the currently available models: (1) establishing tolerance of the mouse host for the human graft; (2) opening hematopoietic niches so that they can be occupied by human cells; and (3) providing necessary support for human hematopoiesis. We then discuss four remaining challenges: (1) human hematopoietic lineages that poorly develop in mice; (2) limited antigen-specific adaptive immunity; (3) absent tolerance of the human immune system for its mouse host; and (4) sub-functional interactions between human immune effectors and target mouse tissues. While major advances are still needed, the current models can already be used to answer specific, clinically-relevant questions and hopefully inform the development of new, life-saving therapies.


Asunto(s)
Inmunidad Adaptativa , Modelos Animales de Enfermedad , Hematopoyesis/inmunología , Trasplante de Células Madre Hematopoyéticas , Células Madre Hematopoyéticas/inmunología , Animales , Xenoinjertos , Humanos , Ratones
19.
Blood ; 137(22): 3015-3026, 2021 06 03.
Artículo en Inglés | MEDLINE | ID: mdl-33684935

RESUMEN

The bone marrow (BM) is responsible for generating and maintaining lifelong output of blood and immune cells. In addition to its key hematopoietic function, the BM acts as an important lymphoid organ, hosting a large variety of mature lymphocyte populations, including B cells, T cells, natural killer T cells, and innate lymphoid cells. Many of these cell types are thought to visit the BM only transiently, but for others, like plasma cells and memory T cells, the BM provides supportive niches that promote their long-term survival. Interestingly, accumulating evidence points toward an important role for mature lymphocytes in the regulation of hematopoietic stem cells (HSCs) and hematopoiesis in health and disease. In this review, we describe the diversity, migration, localization, and function of mature lymphocyte populations in murine and human BM, focusing on their role in immunity and hematopoiesis. We also address how various BM lymphocyte subsets contribute to the development of aplastic anemia and immune thrombocytopenia, illustrating the complexity of these BM disorders and the underlying similarities and differences in their disease pathophysiology. Finally, we summarize the interactions between mature lymphocytes and BM resident cells in HSC transplantation and graft-versus-host disease. A better understanding of the mechanisms by which mature lymphocyte populations regulate BM function will likely improve future therapies for patients with benign and malignant hematologic disorders.


Asunto(s)
Células de la Médula Ósea/inmunología , Enfermedad Injerto contra Huésped , Neoplasias Hematológicas , Células Madre Hematopoyéticas/inmunología , Linfocitos/inmunología , Trombocitopenia , Aloinjertos , Animales , Células de la Médula Ósea/patología , Movimiento Celular/inmunología , Enfermedad Injerto contra Huésped/inmunología , Enfermedad Injerto contra Huésped/patología , Enfermedad Injerto contra Huésped/fisiopatología , Enfermedad Injerto contra Huésped/terapia , Neoplasias Hematológicas/inmunología , Neoplasias Hematológicas/patología , Neoplasias Hematológicas/fisiopatología , Neoplasias Hematológicas/terapia , Hematopoyesis/inmunología , Trasplante de Células Madre Hematopoyéticas , Células Madre Hematopoyéticas/patología , Humanos , Inmunidad Innata , Linfocitos/patología , Ratones , Trombocitopenia/inmunología , Trombocitopenia/patología , Trombocitopenia/fisiopatología , Trombocitopenia/terapia
20.
Signal Transduct Target Ther ; 6(1): 24, 2021 01 20.
Artículo en Inglés | MEDLINE | ID: mdl-33468999

RESUMEN

Hematopoiesis requires finely tuned regulation of gene expression at each stage of development. The regulation of gene transcription involves not only individual transcription factors (TFs) but also transcription complexes (TCs) composed of transcription factor(s) and multisubunit cofactors. In their normal compositions, TCs orchestrate lineage-specific patterns of gene expression and ensure the production of the correct proportions of individual cell lineages during hematopoiesis. The integration of posttranslational and conformational modifications in the chromatin landscape, nucleosomes, histones and interacting components via the cofactor-TF interplay is critical to optimal TF activity. Mutations or translocations of cofactor genes are expected to alter cofactor-TF interactions, which may be causative for the pathogenesis of various hematologic disorders. Blocking TF oncogenic activity in hematologic disorders through targeting cofactors in aberrant complexes has been an exciting therapeutic strategy. In this review, we summarize the current knowledge regarding the models and functions of cofactor-TF interplay in physiological hematopoiesis and highlight their implications in the etiology of hematological malignancies. This review presents a deep insight into the physiological and pathological implications of transcription machinery in the blood system.


Asunto(s)
Neoplasias Hematológicas/inmunología , Hematopoyesis/inmunología , Proteínas de Neoplasias/inmunología , Factores de Transcripción/inmunología , Animales , Humanos
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA