Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 39
Filtrar
1.
FASEB J ; 38(6): e23555, 2024 Mar 31.
Artículo en Inglés | MEDLINE | ID: mdl-38498346

RESUMEN

Dysregulated inflammation-resolution programs are associated with atherosclerosis progression. Resolvins, in part, mediate inflammation-resolution programs. Indeed, Resolvin D2 (RvD2) activates GPR18, a G-protein-coupled receptor, and limits plaque progression, though the cellular targets of RvD2 remain unknown. Here, we developed a humanized GPR18 floxed ("fl/fl") and a myeloid (Lysozyme M Cre) GPR18 knockout (mKO) mouse. We functionally validated this model by assessing efferocytosis in bone marrow-derived macrophages (BMDMs) and found that RvD2 enhanced efferocytosis in the fl/fl, but not in the mKO BMDMs. To understand the functions of RvD2-GPR18 in atherosclerosis, we performed a bone marrow transfer of fl/fl or mKO bone marrow into Ldlr-/- recipients. For these experiments, we treated each genotype with either Vehicle/PBS or RvD2 (25 ng/mouse, 3 times/week for 3 weeks). Myeloid loss of GPR18 resulted in significantly more necrosis, increased cleaved caspase-3+ cells and decreased percentage of Arginase-1+ -Mac2+ cells without a change in overall Mac2+ plaque macrophages, compared with fl/fl➔Ldlr-/- transplanted mice. RvD2 treatment decreased plaque necrosis, the percent of cleaved caspase-3+ cells and increased the percent of Arginase-1+ -Mac2+ cells in fl/fl➔Ldlr-/- mice, but not in the mKO➔Ldlr-/- transplanted mice. These results suggest that GPR18 plays a causal role in limiting atherosclerosis progression and that RvD2's ability to limit plaque necrosis is in part dependent on myeloid GRP18.


Asunto(s)
Arginasa , Aterosclerosis , Ácidos Docosahexaenoicos , Ratones , Animales , Caspasa 3 , Macrófagos , Inflamación , Aterosclerosis/genética , Necrosis , Receptores Acoplados a Proteínas G/genética
2.
Am J Pathol ; 193(12): 1953-1968, 2023 12.
Artículo en Inglés | MEDLINE | ID: mdl-37717941

RESUMEN

Aging is associated with nonresolving inflammation and tissue dysfunction. Resolvin D2 (RvD2) is a proresolving ligand that acts through the G-protein-coupled receptor called GPR18. Unbiased RNA sequencing revealed increased Gpr18 expression in macrophages from old mice, and in livers from elderly humans, which was associated with increased steatosis and fibrosis in middle-aged (MA) and old mice. MA mice that lacked GPR18 on myeloid cells had exacerbated steatosis and hepatic fibrosis, which was associated with a decline in Mac2+ macrophages. Treatment of MA mice with RvD2 reduced steatosis and decreased hepatic fibrosis, correlating with increased Mac2+ macrophages, increased monocyte-derived macrophages, and elevated numbers of monocytes in the liver, blood, and bone marrow. RvD2 acted directly on the bone marrow to increase monocyte-macrophage progenitors. A transplantation assay further demonstrated that bone marrow from old mice facilitated hepatic collagen accumulation in young mice. Transient RvD2 treatment to mice transplanted with bone marrow from old mice prevented hepatic collagen accumulation. Together, this study demonstrates that RvD2-GPR18 signaling controls steatosis and fibrosis and provides a mechanistic-based therapy for promoting liver repair in aging.


Asunto(s)
Médula Ósea , Hígado Graso , Persona de Mediana Edad , Humanos , Ratones , Animales , Anciano , Médula Ósea/metabolismo , Receptores Acoplados a Proteínas G/metabolismo , Envejecimiento , Cirrosis Hepática , Fibrosis , Colágeno/genética , Ratones Endogámicos C57BL
3.
J Immunol ; 207(7): 1812-1823, 2021 10 01.
Artículo en Inglés | MEDLINE | ID: mdl-34462312

RESUMEN

Radiation is associated with tissue damage and increased risk of atherosclerosis, but there are currently no treatments and a very limited mechanistic understanding of how radiation impacts tissue repair mechanisms. We uncovered that radiation significantly delayed temporal resolution programs that were associated with decreased efferocytosis in vivo. Resolvin D1 (RvD1), a known proresolving ligand, promoted swift resolution and restored efferocytosis in sublethally irradiated mice. Irradiated macrophages exhibited several features of senescence, including increased expression of p16INK4A and p21, heightened levels of SA-ß-gal, COX-2, several proinflammatory cytokines/chemokines, and oxidative stress (OS) in vitro, and when transferred to mice, they exacerbated inflammation in vivo. Mechanistically, heightened OS in senescent macrophages led to impairment in their ability to carry out efficient efferocytosis, and treatment with RvD1 reduced OS and improved efferocytosis. Sublethally irradiated Ldlr -/- mice exhibited increased plaque necrosis, p16INK4A cells, and decreased lesional collagen compared with nonirradiated controls, and treatment with RvD1 significantly reduced necrosis and increased lesional collagen. Removal of p16INK4A hematopoietic cells during advanced atherosclerosis with p16-3MR mice reduced plaque necrosis and increased production of key intraplaque-resolving mediators. Our results demonstrate that sublethal radiation drives macrophage senescence and efferocytosis defects and suggest that RvD1 may be a new therapeutic strategy to limit radiation-induced tissue damage.


Asunto(s)
Aterosclerosis/inmunología , Enfermedades Cardiovasculares/inmunología , Ácidos Docosahexaenoicos/metabolismo , Células Madre Hematopoyéticas/fisiología , Macrófagos/inmunología , Traumatismos por Radiación/inmunología , Cicatrización de Heridas/efectos de la radiación , Animales , Aterosclerosis/genética , Células Cultivadas , Senescencia Celular , Ciclooxigenasa 2/metabolismo , Genes p16 , Humanos , Inflamación , Ratones , Ratones Noqueados , Radiación
4.
FASEB J ; 34(1): 597-609, 2020 01.
Artículo en Inglés | MEDLINE | ID: mdl-31914705

RESUMEN

Inflammation-resolution is mediated by the balance between specialized pro-resolving mediators (SPMs) like resolvin D1 (RvD1) and pro-inflammatory factors, like leukotriene B4 (LTB4). A key cellular process of inflammation-resolution is efferocytosis. Aging is associated with defective inflammation-resolution and the accumulation of pro-inflammatory senescent cells (SCs). Therefore, understanding mechanism(s) that underpin this impairment is a critical gap. Here, using a model of hind limb ischemia-reperfusion (I/R) remote lung injury, we present evidence that aging is associated with heightened inflammation, impaired SPM:LT ratio, defective efferocytosis, and a decrease in MerTK levels in injured lungs. Treatment with RvD1 mitigated I/R lung injury in aging, promoted efferocytosis, and prevented the decrease of MerTK in injured lungs from old mice. Old MerTK cleavage-resistant mice (MerTKCR) exhibited less neutrophils or polymorpho nuclear cells infiltration and had improved efferocytosis compared with old WT controls. Mechanistically, macrophages that were treated with conditioned media (CM) from senescent cells had increased MerTK cleavage, impaired efferocytosis, and a defective RvD1:LTB4 ratio. Macrophages from MerTKCR mice were resistant to CM-induced efferocytosis defects and had an improved RvD1:LTB4 ratio. RvD1-stimulated macrophages prevented CM-induced MerTK cleavage and promoted efferocytosis. Together, these data suggest a new mechanism and a potential therapy to promote inflammation-resolution and efferocytosis in aging.


Asunto(s)
Envejecimiento , Ácidos Docosahexaenoicos/farmacología , Inflamación/tratamiento farmacológico , Tirosina Quinasa c-Mer/efectos de los fármacos , Animales , Senescencia Celular/efectos de los fármacos , Inflamación/metabolismo , Macrófagos/efectos de los fármacos , Macrófagos/metabolismo , Masculino , Ratones Endogámicos C57BL , Ratones Transgénicos , Neutrófilos/metabolismo , Peritonitis/tratamiento farmacológico , Fagocitosis/efectos de los fármacos , Proteínas Tirosina Quinasas Receptoras/metabolismo , Daño por Reperfusión/tratamiento farmacológico , Daño por Reperfusión/metabolismo
5.
PLoS Pathog ; 14(8): e1007234, 2018 08.
Artículo en Inglés | MEDLINE | ID: mdl-30080899

RESUMEN

Type I interferons (IFNα/ß) regulate diverse aspects of host defense, but their impact on hematopoietic stem and progenitor cells (HSC/HSPCs) during infection remains unclear. Hematologic impairment can occur in severe infections, thus we sought to investigate the impact of type I IFNs on hematopoiesis in a tick-borne infection with a virulent ehrlichial pathogen that causes shock-like disease. During infection, IFNα/ß induced severe bone marrow (BM) loss, blunted infection-induced emergency myelopoiesis, and reduced phenotypic HSPCs and HSCs. In the absence of type I IFN signaling, BM and splenic hematopoiesis were increased, and HSCs derived from Ifnar1-deficient mice were functionally superior in competitive BM transplants. Type I IFNs impaired hematopoiesis during infection by both limiting HSC/HSPC proliferation and increasing HSPC death. Using mixed BM chimeras we determined that type I IFNs restricted proliferation indirectly, whereas HSPC death occurred via direct IFNαR -mediated signaling. IFNαR-dependent signals resulted in reduced caspase 8 expression and activity, and reduced cleavage of RIPK1 and RIPK3, relative to Ifnar1-deficient mice. RIPK1 antagonism with Necrostatin-1s rescued HSPC and HSC numbers during infection. Early antibiotic treatment is required for mouse survival, however antibiotic-treated survivors had severely reduced HSPCs and HSCs. Combination therapy with antibiotics and Necrostatin-1s improved HSPC and HSC numbers in surviving mice, compared to antibiotic treatment alone. We reveal two mechanisms whereby type I IFNs drive hematopoietic collapse during severe infection: direct sensitization of HSPCs to undergo cell death and enhanced HSC quiescence. Our studies reveal a strategy to ameliorate the type I IFN-dependent loss of HSCs and HSPCs during infection, which may be relevant to other infections wherein type I IFNs cause hematopoietic dysfunction.


Asunto(s)
Ehrlichiosis/patología , Células Madre Hematopoyéticas/fisiología , Interferón Tipo I/fisiología , Choque/patología , Animales , Células de la Médula Ósea/fisiología , Muerte Celular/efectos de los fármacos , Muerte Celular/genética , Diferenciación Celular/efectos de los fármacos , Diferenciación Celular/genética , Proliferación Celular/efectos de los fármacos , Proliferación Celular/genética , Regulación hacia Abajo/genética , Ehrlichia/patogenicidad , Ehrlichiosis/microbiología , Femenino , Hematopoyesis/efectos de los fármacos , Hematopoyesis/genética , Células Madre Hematopoyéticas/efectos de los fármacos , Interferón Tipo I/farmacología , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Proteína Serina-Treonina Quinasas de Interacción con Receptores/genética , Choque/genética , Choque/microbiología
6.
PLoS Pathog ; 12(3): e1005517, 2016 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-27015566

RESUMEN

Inhalation of Francisella tularensis (Ft) causes acute and fatal pneumonia. The lung cytokine milieu favors exponential Ft replication, but the mechanisms underlying acute pathogenesis and death remain unknown. Evaluation of the sequential and systemic host immune response in pulmonary tularemia reveals that in contrast to overwhelming bacterial burden or cytokine production, an overt innate cellular response to Ft drives tissue pathology and host mortality. Lethal infection with Ft elicits medullary and extra-medullary myelopoiesis supporting recruitment of large numbers of immature myeloid cells and MDSC to the lungs. These cells fail to mature and die, leading to subsequent necrotic lung damage, loss of pulmonary function, and host death that is partially dependent upon immature Ly6G+ cells. Acceleration of this process may account for the rapid lethality seen with Ft SchuS4. In contrast, during sub-lethal infection with Ft LVS the pulmonary cellular response is characterized by a predominance of mature neutrophils and monocytes required for protection, suggesting a required threshold for lethal bacterial infection. Further, eliciting a mature phagocyte response provides transient, but dramatic, innate protection against Ft SchuS4. This study reveals that the nature of the myeloid cell response may be the primary determinant of host mortality versus survival following Francisella infection.


Asunto(s)
Francisella tularensis/inmunología , Receptor Toll-Like 2/metabolismo , Tularemia/inmunología , Animales , Citocinas/metabolismo , Humanos , Inflamación , Pulmón/inmunología , Ratones Endogámicos C57BL , Células Mieloides/metabolismo , Neumonía/metabolismo
7.
Haematologica ; 103(9): 1451-1461, 2018 09.
Artículo en Inglés | MEDLINE | ID: mdl-29773597

RESUMEN

Severe aplastic anemia (SAA) results from profound hematopoietic stem cell loss. T cells and interferon gamma (IFNγ) have long been associated with SAA, yet the underlying mechanisms driving hematopoietic stem cell loss remain unknown. Using a mouse model of SAA, we demonstrate that IFNγ-dependent hematopoietic stem cell loss required macrophages. IFNγ was necessary for bone marrow macrophage persistence, despite loss of other myeloid cells and hematopoietic stem cells. Depleting macrophages or abrogating IFNγ signaling specifically in macrophages did not impair T-cell activation or IFNγ production in the bone marrow but rescued hematopoietic stem cells and reduced mortality. Thus, macrophages are not required for induction of IFNγ in SAA and rather act as sensors of IFNγ. Macrophage depletion rescued thrombocytopenia, increased bone marrow megakaryocytes, preserved platelet-primed stem cells, and increased the platelet-repopulating capacity of transplanted hematopoietic stem cells. In addition to the hematopoietic effects, SAA induced loss of non-hematopoietic stromal populations, including podoplanin-positive stromal cells. However, a subset of podoplanin-positive macrophages was increased during disease, and blockade of podoplanin in mice was sufficient to rescue disease. Our data further our understanding of disease pathogenesis, demonstrating a novel role for macrophages as sensors of IFNγ, thus illustrating an important role for the microenvironment in the pathogenesis of SAA.


Asunto(s)
Anemia Aplásica/etiología , Anemia Aplásica/metabolismo , Regulación de la Expresión Génica , Hematopoyesis , Células Madre Hematopoyéticas/metabolismo , Macrófagos/metabolismo , Glicoproteínas de Membrana/genética , Anemia Aplásica/mortalidad , Anemia Aplásica/patología , Animales , Biomarcadores , Médula Ósea/metabolismo , Médula Ósea/patología , Recuento de Células , Ácido Clodrónico/administración & dosificación , Citocinas/metabolismo , Modelos Animales de Enfermedad , Hematopoyesis/efectos de los fármacos , Hematopoyesis/inmunología , Células Madre Hematopoyéticas/efectos de los fármacos , Inmunofenotipificación , Liposomas , Activación de Linfocitos , Glicoproteínas de Membrana/metabolismo , Ratones , Ratones Transgénicos , Modelos Biológicos , Fenotipo , Índice de Severidad de la Enfermedad , Linfocitos T/inmunología , Linfocitos T/metabolismo , Trombocitopenia/genética , Trombocitopenia/metabolismo , Trombocitopenia/patología
8.
Stem Cells ; 33(7): 2294-305, 2015 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-25880153

RESUMEN

Bone marrow (BM) resident macrophages (Mϕs) regulate hematopoietic stem cell (HSC) mobilization; however, their impact on HSC function has not been investigated. We demonstrate that depletion of BM resident Mϕs increases HSC proliferation as well as the pool of quiescent HSCs. At the same time, during bacterial infection where BM resident Mϕs are selectively increased we observe a decrease in HSC numbers. Moreover, strategies that deplete or reduce Mϕs during infection prevent HSC loss and rescue HSC function. We previously found that the transient loss of HSCs during infection is interferon-gamma (IFNγ)-dependent. We now demonstrate that IFNγ signaling specifically in Mϕs is critical for both the diminished HSC pool and maintenance of BM resident Mϕs during infection. In addition to the IFNγ-dependent loss of BM HSC and progenitor cells (HSPCs) during infection, IFNγ reduced circulating HSPC numbers. Importantly, under infection conditions AMD3100 or G-CSF-induced stem cell mobilization was impaired. Taken together, our data show that IFNγ acts on Mϕs, which are a negative regulator of the HSC pool, to drive the loss in BM and peripheral HSCs during infection. Our findings demonstrate that modulating BM resident Mϕ numbers can impact HSC function in vivo, which may be therapeutically useful for hematologic conditions and refinement of HSC transplantation protocols.


Asunto(s)
Células de la Médula Ósea/metabolismo , Células Madre Hematopoyéticas/metabolismo , Interferón gamma/metabolismo , Macrófagos/metabolismo , Animales , Diferenciación Celular , Proliferación Celular , Femenino , Ratones , Ratones Endogámicos C57BL , Transducción de Señal
9.
J Immunol ; 192(4): 1946-53, 2014 Feb 15.
Artículo en Inglés | MEDLINE | ID: mdl-24446520

RESUMEN

Using several tumor models, we demonstrate that mice deficient in Bcl11b in T cells, although having reduced numbers of T cells in the peripheral lymphoid organs, developed significantly less tumors compared with wild-type mice. Bcl11b(-/-) CD4(+) T cells, with elevated TNF-α levels, but not the Bcl11b(-/-) CD8(+) T cells, were required for the reduced tumor burden, as were NK1.1(+) cells, found in increased numbers in Bcl11b(F/F)/CD4-Cre mice. Among NK1.1(+) cells, the NK cell population was predominant in number and was the only population displaying elevated granzyme B levels and increased degranulation, although not increased proliferation. Although the number of myeloid-derived suppressor cells was increased in the lungs with metastatic tumors of Bcl11b(F/F)/CD4-Cre mice, their arginase-1 levels were severely reduced. The increase in NK cell and myeloid-derived suppressor cell numbers was associated with increased bone marrow and splenic hematopoiesis. Finally, the reduced tumor burden, increased numbers of NK cells in the lung, and increased hematopoiesis in Bcl11b(F/F)/CD4-Cre mice were all dependent on TNF-α. Moreover, TNF-α treatment of wild-type mice also reduced the tumor burden and increased hematopoiesis and the numbers and activity of NK cells in the lung. In vitro treatment with TNF-α of lineage-negative hematopoietic progenitors increased NK and myeloid differentiation, further supporting a role of TNF-α in promoting hematopoiesis. These studies reveal a novel role for TNF-α in the antitumor immune response, specifically in stimulating hematopoiesis and increasing the numbers and activity of NK cells.


Asunto(s)
Melanoma/inmunología , Melanoma/patología , Proteínas Represoras/metabolismo , Linfocitos T Reguladores/inmunología , Carga Tumoral/inmunología , Factor de Necrosis Tumoral alfa/metabolismo , Proteínas Supresoras de Tumor/metabolismo , Animales , Antígenos Ly/metabolismo , Arginasa/metabolismo , Linfocitos T CD8-positivos/inmunología , Degranulación de la Célula/inmunología , Proliferación Celular , Eliminación de Gen , Granzimas/metabolismo , Hematopoyesis , Células Asesinas Naturales/inmunología , Neoplasias Pulmonares/inmunología , Recuento de Linfocitos , Ratones , Ratones Transgénicos , Células Mieloides/inmunología , Subfamilia B de Receptores Similares a Lectina de Células NK/metabolismo , Proteínas Represoras/genética , Bazo/inmunología , Linfocitos T Reguladores/metabolismo , Proteínas Supresoras de Tumor/genética , Regulación hacia Arriba
10.
J Immunol ; 190(9): 4725-35, 2013 May 01.
Artículo en Inglés | MEDLINE | ID: mdl-23526822

RESUMEN

Hematopoietic stem and progenitor cell (HSPC) phenotype and function can change in response to infectious challenge. These changes can be mediated by cytokines, IFNs, and pathogen-associated molecules, via TLR, and are thought to promote tailored immune responses for particular pathogens. In this study, we investigated the signals that activate HSPCs during ehrlichiosis, a disease characterized by profound hematopoietic dysfunction in both humans and mice. In a mouse model of ehrlichiosis, we observed that infection-induced proliferation of bone marrow HSPCs was dependent on IFN-γ signaling and was partially dependent on MyD88. However, MyD88 was not required in HSPCs for their expansion during infection, because similar frequencies of MyD88-deficient and wild-type HSPCs proliferated in mixed bone marrow chimeric mice. MyD88-deficient mice exhibited low serum and bone marrow concentration of IFN-γ compared with wild-type mice. We next identified CD4 T cells as the primary cells producing IFN-γ in the bone marrow and demonstrated a nonredundant role for CD4-derived IFN-γ in increased HSPCs. Using mixed bone marrow chimeric mice, we identified a requirement for MyD88 in CD4 T cells for increased T-bet expression, optimal IFN-γ production, and CD4 T cell proliferation. Our data demonstrate an essential role for CD4 T cells in mediating HSPC activation in response to bacterial infection and illustrate a novel role for MyD88 signaling in CD4 T cells in this process. These findings further support the idea that IFN-γ production is essential for HSPC activation and hematopoietic responses to infection.


Asunto(s)
Infecciones Bacterianas/metabolismo , Linfocitos T CD4-Positivos/metabolismo , Células Madre Hematopoyéticas/metabolismo , Interferón gamma/biosíntesis , Factor 88 de Diferenciación Mieloide/metabolismo , Animales , Infecciones Bacterianas/inmunología , Infecciones Bacterianas/microbiología , Médula Ósea/inmunología , Médula Ósea/metabolismo , Médula Ósea/microbiología , Linfocitos T CD4-Positivos/inmunología , Linfocitos T CD4-Positivos/microbiología , Proliferación Celular , Ehrlichia/inmunología , Ehrlichiosis/inmunología , Ehrlichiosis/metabolismo , Ehrlichiosis/microbiología , Células Madre Hematopoyéticas/inmunología , Células Madre Hematopoyéticas/microbiología , Interferón gamma/inmunología , Interferón gamma/metabolismo , Activación de Linfocitos/inmunología , Macrófagos/inmunología , Macrófagos/metabolismo , Macrófagos/microbiología , Ratones , Factor 88 de Diferenciación Mieloide/inmunología , Transducción de Señal/inmunología
11.
Infect Immun ; 82(4): 1698-709, 2014 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-24491580

RESUMEN

Human monocytic ehrlichiosis (HME) is caused by a tick-borne obligate intracellular pathogen of the order Rickettsiales. HME disease can range from mild to a fatal, toxic shock-like syndrome, yet the mechanisms regulating pathogenesis are not well understood. We define a central role for type I interferons (alpha interferon [IFN-α] and IFN-ß) in severe disease in a mouse model of fatal ehrlichiosis caused by Ixodes ovatus Ehrlichia (IOE). IFN-α and IFN-ß were induced by IOE infection but not in response to a less virulent strain, Ehrlichia muris. The major sources of type I IFNs during IOE infection were plasmacytoid dendritic cells and monocytes. Mice lacking the receptor for type I IFNs (Ifnar deficient) or neutralization of IFN-α and IFN-ß resulted in a reduced bacterial burden. Ifnar-deficient mice exhibited significantly increased survival after IOE infection, relative to that of wild-type (WT) mice, that correlated with increased type II IFN (IFN-γ) production. Pathogen-specific antibody responses were also elevated in Ifnar-deficient mice, and this required IFN-γ. Remarkably, increased IFN-γ and IgM were not essential for protection in the absence of type I IFN signaling. The direct effect of type I IFNs on hematopoietic and nonhematopoietic cells was evaluated in bone marrow chimeric mice. We observed that chimeric mice containing Ifnar-deficient hematopoietic cells succumbed to infection early, whereas Ifnar-deficient mice containing WT hematopoietic cells exhibited increased survival, despite having a higher bacterial burden. These data demonstrate that IFN-α receptor signaling in nonhematopoietic cells is important for pathogenesis. Thus, type I IFNs are induced during a rickettsial infection in vivo and promote severe disease.


Asunto(s)
Ehrlichia/patogenicidad , Ehrlichiosis/inmunología , Interferón Tipo I/fisiología , Interferón-alfa/fisiología , Interferón gamma/fisiología , Análisis de Varianza , Animales , Linfocitos T CD8-positivos/metabolismo , Modelos Animales de Enfermedad , Inmunoglobulina M/fisiología , Ratones , Ratones Endogámicos C57BL
12.
Stem Cell Reports ; 19(5): 639-653, 2024 May 14.
Artículo en Inglés | MEDLINE | ID: mdl-38608679

RESUMEN

Sepsis survivors exhibit immune dysfunction, hematological changes, and increased risk of infection. The long-term impacts of sepsis on hematopoiesis were analyzed using a surgical model of murine sepsis, resulting in 50% survival. During acute disease, phenotypic hematopoietic stem and progenitor cells (HSPCs) were reduced in the bone marrow (BM), concomitant with increased myeloid colony-forming units and extramedullary hematopoiesis. Upon recovery, BM HSPCs were increased and exhibited normal function in the context of transplantation. To evaluate hematopoietic responses in sepsis survivors, we treated recovered sham and cecal ligation and puncture mice with a mobilizing regimen of granulocyte colony-stimulating factor (G-CSF) at day 20 post-surgery. Sepsis survivors failed to undergo emergency myelopoiesis and HSPC mobilization in response to G-CSF administration. G-CSF is produced in response to acute infection and injury to expedite the production of innate immune cells; therefore, our findings contribute to a new understanding of how sepsis predisposes to subsequent infection.


Asunto(s)
Factor Estimulante de Colonias de Granulocitos , Movilización de Célula Madre Hematopoyética , Células Madre Hematopoyéticas , Mielopoyesis , Sepsis , Animales , Masculino , Ratones , Modelos Animales de Enfermedad , Factor Estimulante de Colonias de Granulocitos/farmacología , Factor Estimulante de Colonias de Granulocitos/uso terapéutico , Células Madre Hematopoyéticas/citología , Células Madre Hematopoyéticas/efectos de los fármacos , Células Madre Hematopoyéticas/metabolismo , Ratones Endogámicos C57BL , Mielopoyesis/efectos de los fármacos , Sepsis/complicaciones , Sobrevivientes
13.
Cell Death Dis ; 15(5): 324, 2024 May 09.
Artículo en Inglés | MEDLINE | ID: mdl-38724533

RESUMEN

Severe aplastic anemia (SAA) is a rare, fatal disease characterized by severe cytopenias and loss of hematopoietic stem cells (HSCs). Immune-mediated destruction and inflammation are known drivers of SAA, however, the underlying mechanisms driving persistent inflammation are unknown. Current treatments for SAA rely on immunosuppressive therapies or HSC transplantation, however, these treatments are not always effective. Using an established mouse model of SAA, we observed a significant increase in apoptotic cells within the bone marrow (BM) and impaired efferocytosis in SAA mice, relative to radiation controls. Single-cell transcriptomic analysis revealed heterogeneity among BM monocytes and unique populations emerged during SAA characterized by increased inflammatory signatures and significantly increased expression of Sirpa and Cd47. CD47, a "don't eat me" signal, was increased on both live and apoptotic BM cells, concurrent with markedly increased expression of signal regulatory protein alpha (SIRPα) on monocytes. Functionally, SIRPα blockade improved cell clearance and reduced accumulation of CD47-positive apoptotic cells. Lipidomic analysis revealed a reduction in the precursors of specialized pro-resolving lipid mediators (SPMs) and increased prostaglandins in the BM during SAA, indicative of impaired inflammation resolution. Specifically, 18-HEPE, a precursor of E-series resolvins, was significantly reduced in SAA-induced mice relative to radiation controls. Treatment of SAA mice with Resolvin E1 (RvE1) improved efferocytic function, BM cellularity, platelet output, and survival. Our data suggest that impaired efferocytosis and inflammation resolution contributes to SAA progression and demonstrate that SPMs, such as RvE1, offer new and/or complementary treatments for SAA that do not rely on immune suppression.


Asunto(s)
Anemia Aplásica , Antígeno CD47 , Eferocitosis , Ácido Eicosapentaenoico , Animales , Masculino , Ratones , Anemia Aplásica/patología , Antígeno CD47/metabolismo , Antígeno CD47/genética , Modelos Animales de Enfermedad , Eferocitosis/efectos de los fármacos , Ácido Eicosapentaenoico/análogos & derivados , Ácido Eicosapentaenoico/farmacología , Inflamación/patología , Ratones Endogámicos C57BL , Monocitos/metabolismo , Monocitos/efectos de los fármacos , Receptores Inmunológicos/metabolismo , Receptores Inmunológicos/genética
14.
Infect Immun ; 81(12): 4490-7, 2013 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-24082070

RESUMEN

B cell activating factor of the tumor necrosis factor family (BAFF) is an essential survival factor for B cells and has been shown to regulate T cell-independent (TI) IgM production. During Ehrlichia muris infection, TI IgM secretion in the spleen was BAFF dependent, and antibody-mediated BAFF neutralization led to an impairment of IgM-mediated host defense. The failure of TI plasmablasts to secrete IgM was not a consequence of alterations in their generation, survival, or early differentiation, since all occurred normally in infected mice following BAFF neutralization. Gene expression characteristic of plasma cell differentiation was also unaffected by BAFF neutralization in vivo, and except for CD138, plasmablast cell surface marker expression was unaffected. IgM was produced, since it was detected intracellularly, and impaired secretion was not due to a failure to express the IgM secretory exon. Addition of BAFF to plasmablasts in vitro rescued IgM secretion, suggesting that BAFF signaling can directly regulate secretory processes. Our findings indicate that BAFF signaling can modulate TI host defense by acting at a late stage in B cell differentiation, via its regulation of terminal plasmablast differentiation and/or IgM secretion.


Asunto(s)
Factor Activador de Células B/inmunología , Linfocitos B/inmunología , Ehrlichia/inmunología , Ehrlichiosis/inmunología , Inmunoglobulina M/inmunología , Animales , Factor Activador de Células B/antagonistas & inhibidores , Factor Activador de Células B/metabolismo , Diferenciación Celular , Inmunoglobulina M/metabolismo , Activación de Linfocitos/inmunología , Ratones , Ratones Endogámicos C57BL , Células Plasmáticas/inmunología , Células Plasmáticas/metabolismo , Transducción de Señal/inmunología , Sindecano-1/inmunología , Linfocitos T/inmunología
15.
J Immunol ; 186(2): 1011-21, 2011 Jan 15.
Artículo en Inglés | MEDLINE | ID: mdl-21148037

RESUMEN

IgM responses are well known to occur early postinfection and tend to be short-lived, which has suggested that this Ig does not significantly contribute to long-term immunity. In this study, we demonstrate that chronic infection with the intracellular bacterium Ehrlichia muris elicits a protective, long-term IgM response. Moreover, we identified a population of CD138(high)IgM(high) B cells responsible for Ag-specific IgM production in the bone marrow. The IgM-secreting cells, which exhibited characteristics of both plasmablasts and plasma cells, contributed to protection against fatal ehrlichial challenge. Mice deficient in activation-induced cytidine deaminase, which produce only IgM, were protected against fatal ehrlichial challenge infection. The IgM-secreting cells that we have identified were maintained in the bone marrow in the absence of chronic infection, as antibiotic-treated mice remained protected against challenge infection. Our studies identify a cell population that is responsible for the IgM production in the bone marrow, and they highlight a novel role for IgM in the maintenance of long-term immunity during intracellular bacterial infection.


Asunto(s)
Células de la Médula Ósea/inmunología , Células de la Médula Ósea/microbiología , Ehrlichiosis/inmunología , Ehrlichiosis/prevención & control , Inmunoglobulina M/biosíntesis , Líquido Intracelular/inmunología , Células Plasmáticas/inmunología , Células Plasmáticas/microbiología , Animales , Células de la Médula Ósea/metabolismo , Enfermedad Crónica , Ehrlichia/inmunología , Ehrlichiosis/microbiología , Inmunoglobulina M/fisiología , Líquido Intracelular/microbiología , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Ratones Transgénicos , Células Plasmáticas/metabolismo , Células Precursoras de Linfocitos B/inmunología , Células Precursoras de Linfocitos B/metabolismo , Células Precursoras de Linfocitos B/microbiología , Bazo/inmunología , Bazo/metabolismo , Bazo/microbiología , Factores de Tiempo
16.
J Immunol ; 186(2): 1032-43, 2011 Jan 15.
Artículo en Inglés | MEDLINE | ID: mdl-21149601

RESUMEN

Although microbial infections can alter steady-state hematopoiesis, the mechanisms that drive such changes are not well understood. We addressed a role for IFN-γ signaling in infection-induced bone marrow suppression and anemia in a murine model of human monocytic ehrlichiosis, an emerging tick-borne disease. Within the bone marrow of Ehrlichia muris-infected C57BL/6 mice, we observed a reduction in myeloid progenitor cells, as defined both phenotypically and functionally. Infected mice exhibited a concomitant increase in developing myeloid cells within the bone marrow, an increase in the frequency of circulating monocytes, and an increase in splenic myeloid cells. The infection-induced changes in progenitor cell phenotype were critically dependent on IFN-γ, but not IFN-α, signaling. In mice deficient in the IFN-γ signaling pathway, we observed an increase in myeloid progenitor cells and CDllb(lo)Gr1(lo) promyelocytic cells within the bone marrow, as well as reduced frequencies of mature granulocytes and monocytes. Furthermore, E. muris-infected IFN-γR-deficient mice did not exhibit anemia or an increase in circulating monocytes, and they succumbed to infection. Gene transcription studies revealed that IFN-γR-deficient CDllb(lo)Gr1(lo) promyelocytes from E. muris-infected mice exhibited significantly reduced expression of irf-1 and irf-8, both key transcription factors that regulate the differentiation of granulocytes and monocytes. Finally, using mixed bone marrow chimeric mice, we show that IFN-γ-dependent infection-induced myelopoiesis occurs via the direct effect of the cytokine on developing myeloid cells. We propose that, in addition to its many other known roles, IFN-γ acts to control infection by directly promoting the differentiation of myeloid cells that contribute to host defense.


Asunto(s)
Ehrlichiosis/inmunología , Ehrlichiosis/metabolismo , Interferón gamma/fisiología , Líquido Intracelular/microbiología , Células Mieloides/inmunología , Células Mieloides/microbiología , Mielopoyesis/inmunología , Transducción de Señal/inmunología , Animales , Recuento de Células Sanguíneas , Diferenciación Celular/inmunología , Células Cultivadas , Ehrlichia/inmunología , Ehrlichia/patogenicidad , Ehrlichiosis/patología , Células Madre Hematopoyéticas/inmunología , Células Madre Hematopoyéticas/microbiología , Células Madre Hematopoyéticas/patología , Humanos , Inmunofenotipificación , Líquido Intracelular/inmunología , Líquido Intracelular/metabolismo , Ratones , Ratones Congénicos , Ratones Endogámicos C57BL , Ratones Noqueados , Ratones Transgénicos , Células Mieloides/patología
17.
bioRxiv ; 2023 Aug 02.
Artículo en Inglés | MEDLINE | ID: mdl-36909559

RESUMEN

Current treatments for severe aplastic anemia (SAA) rely on hematopoietic stem cell (HSC) transplantation and immunosuppressive therapies, however these treatments are not always effective. While immune-mediated destruction and inflammation are known drivers of SAA, the underlying mechanisms that lead to persistent inflammation are unknown. Using an established mouse model of SAA, we observed a significant increase in apoptotic cells within the bone marrow (BM) and demonstrate impaired efferocytosis in SAA mice, as compared to radiation controls. Single-cell transcriptomic analysis revealed heterogeneity among BM monocytes and unique populations emerged during SAA characterized by increased inflammatory signatures and significantly increased expression of Sirpa and Cd47. CD47, a "don't eat me" signal, was increased on both live and apoptotic BM cells, concurrent with markedly increased expression of signal regulatory protein alpha (SIRPα) on monocytes. Functionally, SIRPα blockade improved cell clearance and reduced accumulation of CD47-positive apoptotic cells. Lipidomic analysis revealed a reduction in the precursors of specialized pro-resolving lipid mediators (SPMs) and increased prostaglandins in the BM during SAA, indicative of impaired inflammation resolution. Specifically, 18-HEPE, a precursor of E-series resolvins, was significantly reduced in SAA-induced mice relative to radiation controls. Treatment of SAA mice with Resolvin E1 (RvE1) improved efferocytic function, BM cellularity, platelet output, and survival. Our data suggest that impaired efferocytosis and inflammation resolution contributes to SAA progression and demonstrate that SPMs, such as RvE1, offer new and/or complementary treatments for SAA that do not rely on immune suppression.

18.
J Neuroimmunol ; 384: 578204, 2023 11 15.
Artículo en Inglés | MEDLINE | ID: mdl-37774553

RESUMEN

Opioids have long been used for clinical pain management, but also have addictive properties that have contributed to the ongoing opioid epidemic. While opioid activation of opioid receptors is well known to contribute to reward and reinforcement, data now also suggest that opioid activation of immune signaling via toll-like receptor 4 (TLR4) may also play a role in addiction-like processes. TLR4 expression is enriched in immune cells, and in the nervous system is primarily expressed in microglia. Microglial phagocytosis is important for developmental, homeostatic, and pathological processes. To examine how morphine impacts microglial phagocytosis, we isolated microglia from adult male and female rat cortex and striatum and plated them in vitro at 10,000 (10K) or 50,000 cells/well densities. Microglia were incubated with neutral fluorescent microbeads to stimulate phagocytosis in the presence of one of four morphine concentrations. We found that the brain region from which microglia are isolated and plating density, but not morphine concentration, impacts cell survival in vitro. We found that 10-12 M morphine, but not higher concentrations, increases phagocytosis in striatal microglia in vitro independent of sex and plating density, while 10-12 M morphine increased phagocytosis in cortical microglia in vitro independent of sex, but contingent on a plating density. Finally, we demonstrate that the effect of 10-12 M morphine in striatal microglia plated at 10 K density is mediated via TLR4, and not µORs. Overall, our data suggest that in rats, a morphine-TLR4 signaling pathway increases phagocytic activity in microglia independent of sex. This may is useful information for better understanding the possible neural outcomes associated with morphine exposures.


Asunto(s)
Microglía , Morfina , Ratas , Masculino , Femenino , Animales , Morfina/farmacología , Microglía/metabolismo , Analgésicos Opioides/farmacología , Receptor Toll-Like 4/metabolismo , Encéfalo/metabolismo
19.
bioRxiv ; 2023 Jan 06.
Artículo en Inglés | MEDLINE | ID: mdl-36711905

RESUMEN

Aging is associated with non-resolving inflammation and tissue dysfunction. Resolvin D2 (RvD2) is a pro-resolving ligand that acts through the G-protein coupled receptor (GPCR) called GRP18. Using an unbiased screen, we report increased Gpr18 expression in macrophages from old mice and in livers from elderly humans that is associated with increased steatosis and fibrosis in middle-aged (MA) and old mice. MA mice that lack GPR18 on myeloid cells had exacerbated steatosis and hepatic fibrosis, which was associated with a decline in Mac2+ macrophages. Treatment of MA mice with RvD2 reduced steatosis and decreased hepatic fibrosis, correlating with increased Mac2+ macrophages, monocyte-derived macrophages and elevated numbers of monocytes in the liver, blood, and bone marrow. RvD2 acted directly upon the bone marrow to increase monocyte-macrophage progenitors. Using a transplantation assay we further demonstrated that bone marrow from old mice facilitated hepatic collagen accumulation in young mice, and transient RvD2 treatment to mice transplanted with bone marrow from old mice prevented hepatic collagen accumulation. Together, our study demonstrates that RvD2-GPR18 signaling controls steatosis and fibrosis and provides a mechanistic-based therapy for promoting liver repair in aging.

20.
J Immunol ; 184(9): 5085-93, 2010 May 01.
Artículo en Inglés | MEDLINE | ID: mdl-20351185

RESUMEN

Germinal centers (GCs) are specialized microenvironments in secondary lymphoid organs that facilitate the development of high-affinity, isotype-switched Abs, and immunological memory; consequently, many infections require GC-derived IgG for pathogen clearance. Although Ehrlichia muris infection elicits a robust expansion of splenic, IgM-secreting plasmablasts, we detected only very low frequencies of isotype-switched IgG-secreting cells in mouse spleens, until at least 3 wk postinfection. Instead, Ag-specific IgG was produced in lymph nodes, where it required CD4 T cell help. Consistent with these findings, organized GCs and phenotypically defined splenic GC B cells were found in lymph nodes, but not spleens. Ehrlichial infection also inhibited spleen IgG responses against a coadministered T cell-dependent Ag, hapten 4-hydroxy-3-nitrophenyl acetyl (NP)-conjugated chicken gamma globulin in alum. NP-specific B cells failed to undergo expansion and differentiation into GC B cells in the spleen, Ab titers were reduced, and splenic IgG production was inhibited nearly 10-fold when the Ag was administered during infection. Our data provide a mechanism whereby an intracellular bacterial infection can compromise local immunity to coinfecting pathogens or antigenic challenge.


Asunto(s)
Anticuerpos Antibacterianos/metabolismo , Ehrlichia/inmunología , Ehrlichiosis/inmunología , Centro Germinal/inmunología , Inmunoglobulina G/metabolismo , Terapia de Inmunosupresión , Líquido Intracelular/inmunología , Líquido Intracelular/microbiología , Animales , Anticuerpos Antibacterianos/biosíntesis , Anticuerpos Antibacterianos/sangre , Linfocitos T CD4-Positivos/inmunología , Linfocitos T CD4-Positivos/microbiología , Linfocitos T CD4-Positivos/patología , Ehrlichiosis/microbiología , Ehrlichiosis/patología , Epítopos de Linfocito T/inmunología , Centro Germinal/microbiología , Centro Germinal/patología , Inmunoglobulina G/biosíntesis , Inmunoglobulina G/sangre , Inmunoglobulina M/biosíntesis , Líquido Intracelular/metabolismo , Ganglios Linfáticos/inmunología , Ganglios Linfáticos/microbiología , Ganglios Linfáticos/patología , Ratones , Ratones Endogámicos BALB C , Ratones Endogámicos C57BL , Ratones Noqueados , Ratones Transgénicos , Bazo/inmunología , Bazo/microbiología , Bazo/patología
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA