Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 39
Filtrar
1.
Lancet Respir Med ; 12(4): 305-322, 2024 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-38142698

RESUMEN

Sepsis is characterised by a dysregulated host immune response to infection. Despite recognition of its significance, immune status monitoring is not implemented in clinical practice due in part to the current absence of direct therapeutic implications. Technological advances in immunological profiling could enhance our understanding of immune dysregulation and facilitate integration into clinical practice. In this Review, we provide an overview of the current state of immune profiling in sepsis, including its use, current challenges, and opportunities for progress. We highlight the important role of immunological biomarkers in facilitating predictive enrichment in current and future treatment scenarios. We propose that multiple immune and non-immune-related parameters, including clinical and microbiological data, be integrated into diagnostic and predictive combitypes, with the aid of machine learning and artificial intelligence techniques. These combitypes could form the basis of workable algorithms to guide clinical decisions that make precision medicine in sepsis a reality and improve patient outcomes.


Asunto(s)
Medicina de Precisión , Sepsis , Humanos , Medicina de Precisión/métodos , Inteligencia Artificial , Objetivos , Algoritmos , Sepsis/diagnóstico , Sepsis/terapia
2.
Shock ; 59(3S Suppl 1): 21-25, 2023 03 01.
Artículo en Inglés | MEDLINE | ID: mdl-36867758

RESUMEN

ABSTRACT: The unacceptable high mortality of severe infections and sepsis led over the years to understand the need for adjunctive immunotherapy to modulate the dysregulated host response of the host. However, not all patients should receive the same type of treatment. The immune function may largely differ from one patient to the other. The principles of precision medicine require that some biomarker is used to capture the immune function of the host and guide the best candidate therapy. This is the approach of the ImmunoSep randomized clinical trial (NCT04990232) where patients are allocated to treatment with anakinra or recombinant interferon gamma tailored to immune signs of macrophage activation-like syndrome and immunoparalysis respectively. ImmunoSep is a first-in-class paradigm of precision medicine for sepsis. Other approaches need to consider classification by sepsis endotypes, targeting T cell and application of stem cells. Basic principle for any trial to be successful is the delivery of appropriate antimicrobial therapy as standard-of-care taking into consideration not just the likelihood for resistant pathogens but also the pharmacokinetic/pharmacodynamic mode of action of the administered antimicrobial.


Asunto(s)
Sepsis , Choque , Humanos , Inmunoterapia , Interferón gamma/uso terapéutico , Activación de Macrófagos , Medicina de Precisión , Sepsis/tratamiento farmacológico , Choque/tratamiento farmacológico , Ensayos Clínicos Controlados Aleatorios como Asunto , Proteína Antagonista del Receptor de Interleucina 1/uso terapéutico
3.
Front Immunol ; 13: 945409, 2022.
Artículo en Inglés | MEDLINE | ID: mdl-36148245

RESUMEN

Sepsis is associated with profound immune dysregulation that increases the risk for life-threatening secondary infections: Dendritic cells (DCs) undergo functional reprogramming due to yet unknown changes during differentiation in the bone marrow (BM). In parallel, lymphopenia and exhaustion of T lymphocytes interfere with antigen-specific adaptive immunity. We hypothesized that there exists a link between T cells and the modulation of DC differentiation in the BM during murine polymicrobial sepsis. Sepsis was induced by cecal ligation and puncture (CLP), a model for human bacterial sepsis. At different time points after CLP, the BM and spleen were analyzed in terms of T-cell subpopulations, activation, and Interferon (IFN)-γ synthesis as well as the number of pre-DCs. BM-derived DCs were generated in vitro. We observed that naïve and virtual memory CD8+ T cells, but not CD4+ T cells, were activated in an antigen-independent manner and accumulated in the BM early after CLP, whereas lymphopenia was evident in the spleen. The number of pre-DCs strongly declined during acute sepsis in the BM and almost recovered by day 4 after CLP, which required the presence of CD8+ T cells. Adoptive transfer experiments and in vitro studies with purified T cells revealed that Toll-like receptor 2 (TLR2) signaling in CD8+ T cells suppressed their capacity to secrete IFN-γ and was sufficient to change the transcriptome of the BM during sepsis. Moreover, the diminished IFN-γ production of CD8+ T cells favored the differentiation of DCs with increased production of the immune-activating cytokine Interleukin (IL)-12. These data identify a novel role of CD8+ T cells in the BM during sepsis as they sense TLR2 ligands and control the number and function of de novo differentiating DCs.


Asunto(s)
Linfopenia , Sepsis , Animales , Antígenos , Médula Ósea , Linfocitos T CD8-positivos , Diferenciación Celular , Citocinas , Células Dendríticas , Humanos , Interferón gamma , Interleucina-12 , Ratones , Receptor Toll-Like 2
4.
Immunohorizons ; 5(5): 298-306, 2021 05 12.
Artículo en Inglés | MEDLINE | ID: mdl-33980662

RESUMEN

Acute major tissue injury induces immune dysregulation that is characterized by the development of systemic sterile inflammation and an increased risk for opportunistic infections. Although the contribution of the innate immune system has been examined in detail, research on the impact of acute sterile tissue damage on the T cell compartment remains limited. In the current study, we used a clinically relevant mouse model for traumatic skeletal muscle injury to investigate the impact of sterile tissue damage on diverse subpopulations of CD4+ Th and CD8+ cytotoxic T cells in systemic and local lymphoid organs. For the first time, to our knowledge, we provide evidence that injury selectively induced the expression of the activation marker CD69 on naive and central/virtual memory CD8+ T cells in the lymph nodes but not in the spleen of male mice. CD4+ Th cells remained unaffected in both organs. The activation of CD8+ T cells was dependent on signaling through TLR4. Within a few hours, injury triggered the expression of IL-12 in the lymph nodes in a TLR4-dependent manner. Blocking of IL-12 prevented the activation of naive and central memory CD8+ T cells after injury. Thus, early after traumatic tissue damage, TLR4 transactivates naive and central/virtual memory CD8+ T cells through innate cytokines in local lymph nodes, where they might modulate forthcoming local immune responses.


Asunto(s)
Linfocitos T CD8-positivos/inmunología , Citocinas/inmunología , Músculo Esquelético/lesiones , Receptor Toll-Like 4/genética , Animales , Citocinas/genética , Modelos Animales de Enfermedad , Ganglios Linfáticos/citología , Masculino , Ratones , Ratones Endogámicos BALB C , Bazo/citología
5.
Lancet Respir Med ; 9(6): 622-642, 2021 06.
Artículo en Inglés | MEDLINE | ID: mdl-33965003

RESUMEN

The zoonotic SARS-CoV-2 virus that causes COVID-19 continues to spread worldwide, with devastating consequences. While the medical community has gained insight into the epidemiology of COVID-19, important questions remain about the clinical complexities and underlying mechanisms of disease phenotypes. Severe COVID-19 most commonly involves respiratory manifestations, although other systems are also affected, and acute disease is often followed by protracted complications. Such complex manifestations suggest that SARS-CoV-2 dysregulates the host response, triggering wide-ranging immuno-inflammatory, thrombotic, and parenchymal derangements. We review the intricacies of COVID-19 pathophysiology, its various phenotypes, and the anti-SARS-CoV-2 host response at the humoral and cellular levels. Some similarities exist between COVID-19 and respiratory failure of other origins, but evidence for many distinctive mechanistic features indicates that COVID-19 constitutes a new disease entity, with emerging data suggesting involvement of an endotheliopathy-centred pathophysiology. Further research, combining basic and clinical studies, is needed to advance understanding of pathophysiological mechanisms and to characterise immuno-inflammatory derangements across the range of phenotypes to enable optimum care for patients with COVID-19.


Asunto(s)
COVID-19 , Insuficiencia Multiorgánica , SARS-CoV-2/patogenicidad , COVID-19/inmunología , COVID-19/fisiopatología , Endotelio/fisiopatología , Humanos , Inmunidad , Insuficiencia Multiorgánica/etiología , Insuficiencia Multiorgánica/fisiopatología , Gravedad del Paciente , Índice de Severidad de la Enfermedad
6.
EBioMedicine ; 66: 103291, 2021 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-33813139

RESUMEN

Many milestones in medical history rest on animal modeling of human diseases. The SARS-CoV-2 pandemic has evoked a tremendous investigative effort primarily centered on clinical studies. However, several animal SARS-CoV-2/COVID-19 models have been developed and pre-clinical findings aimed at supporting clinical evidence rapidly emerge. In this review, we characterize the existing animal models exposing their relevance and limitations as well as outline their utility in COVID-19 drug and vaccine development. Concurrently, we summarize the status of clinical trial research and discuss the novel tactics utilized in the largest multi-center trials aiming to accelerate generation of reliable results that may subsequently shape COVID-19 clinical treatment practices. We also highlight areas of improvement for animal studies in order to elevate their translational utility. In pandemics, to optimize the use of strained resources in a short time-frame, optimizing and strengthening the synergy between the preclinical and clinical domains is pivotal.


Asunto(s)
Tratamiento Farmacológico de COVID-19 , Vacunas contra la COVID-19 , COVID-19/etiología , Modelos Animales de Enfermedad , SARS-CoV-2/genética , Factores de Edad , Animales , Antivirales/farmacología , COVID-19/fisiopatología , COVID-19/terapia , Vacunas contra la COVID-19/farmacología , Ensayos Clínicos como Asunto , Cricetinae , Hurones , Humanos , Ratones , Mutación , Primates
7.
Life (Basel) ; 12(1)2021 Dec 22.
Artículo en Inglés | MEDLINE | ID: mdl-35054405

RESUMEN

Major traumatic and surgical injury increase the risk for infectious complications due to immune dysregulation. Upon stimulation with interleukin (IL) 12 by monocyte/macrophages, natural killer (NK) cells release interferon (IFN) γ that supports the elimination of the pathogen. In the present study, we investigated the impact of invasive spine surgery on the relationship between monocytes and NK cells upon exposure to Staphylococcus aureus. Mononuclear cells and serum were isolated from peripheral blood of patients before and up to 8 d after surgery and stimulated with inactivated S. aureus bacteria. NK cell and monocyte function were determined by flow cytometry. NK cells continuously lost their ability to produce IFN-γ during the first week after surgery independently from monocyte-derived IL-12 secretion. IFN-γ synthesis was minimal on day 8 and was associated with decreased expression of the IL-12 receptor and activation of transcription factors required for IFNG gene transcription. Addition of recombinant IL-12 could at least partially restore NK cell function. Pre-operative levels of growth/differentiation factor (GDF) 15 in the serum correlated with the extent of NK cell suppression and with hospitalization. Thus, NK cell suppression after major surgery might represent a therapeutic target to improve the immune defense against opportunistic infections.

8.
Front Immunol ; 11: 1200, 2020.
Artículo en Inglés | MEDLINE | ID: mdl-32670280

RESUMEN

Major trauma-induced tissue injury causes a dysregulation of the immune system. Severe systemic inflammation occurs early after the insult. Later on, an enhanced risk for life-threatening opportunistic infections develops that culminates at the end of the first week after trauma. CD56bright Natural killer (NK) cells play a key role in the defense against infection due to their rapid release of Interferon (IFN) γ in response to Interleukin (IL) 12. NK cells are impaired in IFN-γ synthesis after severe injury due to a disturbed IL-12/IFN-γ axis. Thereby, a circulating factor mediates extrinsic suppression of NK cells. Yet unknown cell-intrinsic mechanisms manifest by day 8 after trauma and render NK cells unresponsive to stimulatory cytokines. In the present study, we investigated the origin of such late NK cell-intrinsic suppression after major trauma. Peripheral blood mononuclear cells (PBMC) were isolated from patients 8 day after severe injury and from healthy control subjects and were stimulated with inactivated Staphylococcus aureus. The expression of diverse cytokine receptors, intracellular signaling molecules, and the secretion of IFN-γ by CD56bright NK cells were examined. After stimulation with S. aureus, NK cells from patients expressed enhanced levels of c-kit/CD117 that inversely correlated with IFN-γ synthesis and IL-12 receptor (IL-12R) ß2 expression. Supplementation with IL-15 and inhibition of the transforming growth factor receptor (TGF-ßR) I reduced CD117 expression and increased the level of IL-12Rß2 and IFN-γ. NK cells from patients showed reduced phosphorylation of mammalian target of rapamycin (mTOR). Addition of IL-15 at least partly restored mTOR phosphorylation and increased IL-12Rß2 expression. The reduced mTOR phosphorylation after severe injury was cell-intrinsic as it was not induced by serum factors. Inhibition of mTOR in purified NK cells from healthy donors by rapamycin decreased the synthesis of IFN-γ. Thus, impaired mTOR phosphorylation in response to a microbial challenge contributes to the cell-intrinsic mechanisms that underlie NK cell dysregulation after trauma. Restoration of the mTOR phosphorylation capacity along with inhibition of the TGF-ßRI signaling in NK cells after severe injury might improve the immune defense against opportunistic infections.


Asunto(s)
Células Asesinas Naturales/inmunología , Proteínas Proto-Oncogénicas c-kit/inmunología , Serina-Treonina Quinasas TOR/inmunología , Heridas y Lesiones/inmunología , Humanos , Células Asesinas Naturales/metabolismo , Proteínas Proto-Oncogénicas c-kit/metabolismo , Serina-Treonina Quinasas TOR/metabolismo
9.
Shock ; 54(4): 416-437, 2020 10.
Artículo en Inglés | MEDLINE | ID: mdl-32433217

RESUMEN

Approximately 3 billion people around the world have gone into some form of social separation to mitigate the current severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) pandemic. The uncontrolled influx of patients in need of emergency care has rapidly brought several national health systems to near-collapse with deadly consequences to those afflicted by Coronavirus Disease 2019 (COVID-19) and other critical diseases associated with COVID-19. Solid scientific evidence regarding SARS-CoV-2/COVID-19 remains scarce; there is an urgent need to expand our understanding of the SARS-CoV-2 pathophysiology to facilitate precise and targeted treatments. The capacity for rapid information dissemination has emerged as a double-edged sword; the existing gap of high-quality data is frequently filled by anecdotal reports, contradictory statements, and misinformation. This review addresses several important aspects unique to the SARS-CoV-2/COVID-19 pandemic highlighting the most relevant knowledge gaps and existing windows-of-opportunity. Specifically, focus is given on SARS-CoV-2 immunopathogenesis in the context of experimental therapies and preclinical evidence and their applicability in supporting efficacious clinical trial planning. The review discusses the existing challenges of SARS-CoV-2 diagnostics and the potential application of translational technology for epidemiological predictions, patient monitoring, and treatment decision-making in COVID-19. Furthermore, solutions for enhancing international strategies in translational research, cooperative networks, and regulatory partnerships are contemplated.


Asunto(s)
Betacoronavirus , Técnicas de Laboratorio Clínico , Infecciones por Coronavirus/diagnóstico , Infecciones por Coronavirus/terapia , Neumonía Viral/diagnóstico , Neumonía Viral/terapia , COVID-19 , Prueba de COVID-19 , Ensayos Clínicos como Asunto , Infecciones por Coronavirus/tratamiento farmacológico , Infecciones por Coronavirus/transmisión , Humanos , Pandemias , Neumonía Viral/transmisión , SARS-CoV-2 , Tratamiento Farmacológico de COVID-19
10.
Stem Cell Res Ther ; 10(1): 338, 2019 11 21.
Artículo en Inglés | MEDLINE | ID: mdl-31753037

RESUMEN

BACKGROUND: During joint replacement, surgical vacuum suction guarantees a sufficient overview on the situs. We assume high concentrations of mesenchymal stromal cells (MSCs) on surgical vacuum filters. We compared the in vitro proliferative and differentiation potency of cells from the following: (i) bone marrow (BM), (ii) cancellous bone (CB), (iii) vacuum filter (VF), and (iv) cell saver filtrate reservoir (SF) in 32 patients undergoing elective total hip replacement. METHODS: Mononuclear cells (MNC) were isolated, and cell proliferation and colony-forming units (CFU) were measured. Adherent cells were characterized by flow cytometry for MSC surface markers. Cells were incubated with osteogenic, adipogenic, and chondrogenic stimuli. Cells were cytochemically stained and osteoblastic expression (RUNX-2, ALP, and BMP-2) investigated via qPCR. RESULTS: Dependent on the source, initial MNC amount as well as CFU number was significantly different whereas generation time did not vary significantly. CFU numbers from VF were superior to those from SR, BM, and CB. The resulting amount of MSC from the respective source was highest in the vacuum filter followed by reservoir, aspirate, and cancellous bone. Cells from all groups could be differentiated into the three mesenchymal lines demonstrating their stemness nature. However, gene expression of osteoblastic markers did not differ significantly between the groups. CONCLUSION: We conclude that surgical vacuum filters are able to concentrate tissue with relevant amounts of MSCs. A new potent source of autologous regeneration material with clinical significance is identified. Further clinical studies have to elucidate the regenerative potential of this material in an autologous setting.


Asunto(s)
Células de la Médula Ósea/metabolismo , Diferenciación Celular , Proliferación Celular , Separación Celular , Células Madre Mesenquimatosas/metabolismo , Anciano , Células de la Médula Ósea/citología , Femenino , Humanos , Masculino , Células Madre Mesenquimatosas/citología , Persona de Mediana Edad , Estudios Prospectivos
11.
Lancet Infect Dis ; 19(12): e422-e436, 2019 12.
Artículo en Inglés | MEDLINE | ID: mdl-31630991

RESUMEN

Increasing evidence supports a central role of the immune system in sepsis, but the current view of how sepsis affects immunity, and vice versa, is still rudimentary. The European Group on Immunology of Sepsis has identified major gaps that should be addressed with high priority, such as understanding how immunological alterations predispose to sepsis, key aspects of the immunopathological events during sepsis, and the long-term consequences of sepsis on patient's immunity. We discuss major unmet topics in those three categories, including the role of key immune cells, the cause of lymphopenia, organ-specific immunology, the dynamics of sepsis-associated immunological alterations, the role of the microbiome, the standardisation of immunological tests, the development of better animal models, and the opportunities offered by immunotherapy. Addressing these gaps should help us to better understand sepsis physiopathology, offering translational opportunities to improve its prevention, diagnosis, and care.


Asunto(s)
Susceptibilidad a Enfermedades/inmunología , Interacciones Huésped-Patógeno/inmunología , Sepsis/etiología , Inmunidad Adaptativa , Animales , Biomarcadores , Manejo de la Enfermedad , Humanos , Sistema Inmunológico/inmunología , Sistema Inmunológico/metabolismo , Inmunidad Innata , Medicina de Precisión/métodos , Factores de Riesgo , Sepsis/diagnóstico , Sepsis/terapia , Investigación Biomédica Traslacional
12.
EBioMedicine ; 43: 380-391, 2019 May.
Artículo en Inglés | MEDLINE | ID: mdl-30992245

RESUMEN

BACKGROUND: Systemic inflammation induced by sterile or infectious insults is associated with an enhanced susceptibility to life-threatening opportunistic, mostly bacterial, infections due to unknown pathogenesis. Natural killer (NK) cells contribute to the defence against bacterial infections through the release of Interferon (IFN) γ in response to Interleukin (IL) 12. Considering the relevance of NK cells in the immune defence we investigated whether the function of NK cells is disturbed in patients suffering from serious systemic inflammation. METHODS: NK cells from severely injured patients were analysed from the first day after the initial inflammatory insult until the day of discharge in terms of IL-12 receptor signalling and IFN-γ synthesis. FINDINGS: During systemic inflammation, the expression of the IL-12 receptor ß2 chain, phosphorylation of signal transducer and activation 4, and IFN-γ production on/in NK cells was impaired upon exposure to Staphylococcus aureus. The profound suppression of NK cells developed within 24 h after the initial insult and persisted for several weeks. NK cells displayed signs of exhaustion. Extrinsic changes were mediated by the early and long-lasting presence of growth/differentiation factor (GDF) 15 in the circulation that signalled through the transforming growth factor ß receptor I and activated Smad1/5. Moreover, the concentration of GDF-15 in the serum inversely correlated with the IL-12 receptor ß2 expression on NK cells and was enhanced in patients who later acquired septic complications. INTERPRETATION: GDF-15 is associated with the development of NK cell dysfunction during systemic inflammation and might represent a novel target to prevent nosocomial infections. FUND: The study was supported by the Department of Orthopaedics and Trauma Surgery, University Hospital Essen.


Asunto(s)
Antígeno CD56/metabolismo , Infección Hospitalaria/etiología , Infección Hospitalaria/metabolismo , Factor 15 de Diferenciación de Crecimiento/sangre , Células Asesinas Naturales/inmunología , Células Asesinas Naturales/metabolismo , Adulto , Biomarcadores , Comorbilidad , Infección Hospitalaria/sangre , Infección Hospitalaria/diagnóstico , Femenino , Humanos , Inmunofenotipificación , Mediadores de Inflamación/metabolismo , Interferón gamma/metabolismo , Interleucina-12/metabolismo , Masculino , Persona de Mediana Edad , Fosforilación , Receptores de Interleucina-12/metabolismo , Factor de Transcripción STAT4/metabolismo , Índice de Severidad de la Enfermedad , Transducción de Señal , Síndrome de Respuesta Inflamatoria Sistémica/sangre , Síndrome de Respuesta Inflamatoria Sistémica/etiología , Síndrome de Respuesta Inflamatoria Sistémica/metabolismo
13.
Front Immunol ; 8: 1622, 2017.
Artículo en Inglés | MEDLINE | ID: mdl-29218051

RESUMEN

Sepsis is the dysregulated response of the host to systemic, mostly bacterial infection, and is associated with an enhanced susceptibility to life-threatening opportunistic infections. During polymicrobial sepsis, dendritic cells (DCs) secrete enhanced levels of interleukin (IL) 10 due to an altered differentiation in the bone marrow and contribute to the development of immunosuppression. We investigated the origin of the altered DC differentiation using murine cecal ligation and puncture (CLP), a model for human polymicrobial sepsis. Bone marrow cells (BMC) were isolated after sham or CLP operation, the cellular composition was analyzed, and bone marrow-derived DCs (BMDCs) were generated in vitro. From 24 h on after CLP, BMC gave rise to BMDC that released enhanced levels of IL-10. In parallel, a population of CD11chiMHCII+CD4+ DCs expanded in the bone marrow in a MyD88-dependent manner. Prior depletion of the CD11chiMHCII+CD4+ DCs from BMC in vitro reversed the increased IL-10 secretion of subsequently differentiating BMDC. The expansion of the CD11chiMHCII+CD4+ DC population in the bone marrow after CLP required the function of sphingosine 1-phosphate receptors and C-C chemokine receptor (CCR) 2, the receptor for C-C chemokine ligand (CCL) 2, but was not associated with monocyte mobilization. CD11chiMHCII+CD4+ DCs were identified as plasmacytoid DCs (pDCs) that had acquired an activated phenotype according to their increased expression of MHC class II and CD86. A redistribution of CD4+ pDCs from MHC class II- to MHC class II+ cells concomitant with enhanced expression of CD11c finally led to the rise in the number of CD11chiMHCII+CD4+ DCs. Enhanced levels of CCL2 were found in the bone marrow of septic mice and the inhibition of CCR2 dampened the expression of CD86 on CD4+ pDCs after CLP in vitro. Depletion of pDCs reversed the bias of splenic DCs toward increased IL-10 synthesis after CLP in vivo. Thus, during polymicrobial sepsis, CD4+ pDCs are activated in the bone marrow and induce functional reprogramming of differentiating BMDC toward an immunosuppressive phenotype.

14.
Stem Cell Reports ; 9(3): 985-998, 2017 09 12.
Artículo en Inglés | MEDLINE | ID: mdl-28781075

RESUMEN

The interaction of mesenchymal stromal cells (MSCs) with natural killer (NK) cells is traditionally thought of as a static inhibitory model, whereby resting MSCs inhibit NK cell effector function. Here, we use a dynamic in vitro system of poly(I:C) stimulation to model the interaction of NK cells and tissue-resident MSCs in the context of infection or tissue injury. The experiments suggest a time-dependent system of regulation and feedback, where, at early time points, activated MSCs secrete type I interferon to enhance NK cell effector function, while at later time points TGF-ß and IL-6 limit NK cell effector function and terminate inflammatory responses by induction of a regulatory senescent-like NK cell phenotype. Importantly, feedback of these regulatory NK cells to MSCs promotes survival, proliferation, and pro-angiogenic properties. Our data provide additional insight into the interaction of stromal cells and innate immune cells and suggest a model of time-dependent MSC polarization and licensing.


Asunto(s)
Células Asesinas Naturales/citología , Células Madre Mesenquimatosas/citología , Regeneración , Apoptosis/efectos de los fármacos , Células de la Médula Ósea/citología , Células de la Médula Ósea/efectos de los fármacos , Células de la Médula Ósea/metabolismo , Comunicación Celular/efectos de los fármacos , Diferenciación Celular/efectos de los fármacos , Proliferación Celular/efectos de los fármacos , Supervivencia Celular/efectos de los fármacos , Senescencia Celular/efectos de los fármacos , Citotoxicidad Inmunológica/efectos de los fármacos , Humanos , Inflamación/patología , Interleucina-6/metabolismo , Células Asesinas Naturales/efectos de los fármacos , Células Asesinas Naturales/metabolismo , Activación de Linfocitos/efectos de los fármacos , Células Madre Mesenquimatosas/efectos de los fármacos , Células Madre Mesenquimatosas/metabolismo , Mucosa Nasal/citología , Fenotipo , Poli I-C/farmacología , Receptores CXCR4/metabolismo , Regeneración/efectos de los fármacos , Factores de Tiempo , Factor de Crecimiento Transformador beta/metabolismo , Cicatrización de Heridas/efectos de los fármacos
15.
Stem Cell Res Ther ; 7(1): 88, 2016 Jul 07.
Artículo en Inglés | MEDLINE | ID: mdl-27388156

RESUMEN

BACKGROUND: The suppressive effect of mesenchymal stromal/stem cells (MSCs) on diverse immune cells is well known, but it is unclear whether MSCs additionally possess immunostimulatory properties. We investigated the impact of human MSCs on the responsiveness of primary natural killer (NK) cells in terms of cytokine secretion. METHODS: Human MSCs were generated from bone marrow and nasal mucosa. NK cells were isolated from peripheral blood of healthy volunteers or of immunocompromised patients after severe injury. NK cells were cultured with MSCs or with MSC-derived conditioned media in the absence or presence of IL-12 and IL-18. C-C chemokine receptor (CCR) 2, C-C chemokine ligand (CCL) 2, and the interferon (IFN)-γ receptor was blocked by specific inhibitors or antibodies. The synthesis of IFN-γ and CCL2 was determined. RESULTS: In the absence of exogenous cytokines, trace amounts of NK cell-derived IFN-γ licensed MSCs for enhanced synthesis of CCL2. In turn, MSCs primed NK cells for increased release of IFN-γ in response to IL-12 and IL-18. Priming of NK cells by MSCs occurred in a cell-cell contact-independent manner and was impaired by inhibition of the CCR2, the receptor of CCL2, on NK cells. CD56(bright) NK cells expressed higher levels of CCR2 and were more sensitive to CCL2-mediated priming by MSCs and by recombinant CCR2 ligands than cytotoxic CD56(dim) NK cells. NK cells from severely injured patients were impaired in cytokine-induced IFN-γ synthesis. Co-culture with MSCs or with conditioned media from MSCs and MSC/NK cell co-cultures from healthy donors improved the IFN-γ production of the patients' NK cells in a CCR2-dependent manner. CONCLUSIONS: A positive feedback loop driven by NK cell-derived IFN-γ and MSC-derived CCL2 increases the inflammatory response of cytokine-stimulated NK cells not only from healthy donors but also from immunocompromised patients. Therapeutic application of MSCs or their soluble factors might thus improve the NK function after severe injury.


Asunto(s)
Huésped Inmunocomprometido , Inmunomodulación/efectos de los fármacos , Células Asesinas Naturales/inmunología , Células Madre Mesenquimatosas/inmunología , Traumatismo Múltiple/inmunología , Adulto , Anticuerpos/farmacología , Comunicación Celular/efectos de los fármacos , Quimiocina CCL2/antagonistas & inhibidores , Quimiocina CCL2/genética , Quimiocina CCL2/inmunología , Técnicas de Cocultivo , Medios de Cultivo Condicionados/farmacología , Femenino , Humanos , Interferón gamma/genética , Interferón gamma/inmunología , Interleucina-12/farmacología , Interleucina-18/farmacología , Células Asesinas Naturales/citología , Células Asesinas Naturales/efectos de los fármacos , Masculino , Células Madre Mesenquimatosas/citología , Células Madre Mesenquimatosas/efectos de los fármacos , Persona de Mediana Edad , Traumatismo Múltiple/patología , Receptores CCR2/antagonistas & inhibidores , Receptores CCR2/genética , Receptores CCR2/inmunología , Receptores de Interferón/genética , Receptores de Interferón/inmunología , Índices de Gravedad del Trauma , Receptor de Interferón gamma
16.
PLoS One ; 11(5): e0155870, 2016.
Artículo en Inglés | MEDLINE | ID: mdl-27196728

RESUMEN

Skeletal muscle injury causes a local sterile inflammatory response. In parallel, a state of immunosuppression develops distal to the site of tissue damage. Granulocytes and monocytes that are rapidly recruited to the site of injury contribute to tissue regeneration. In this study we used a mouse model of traumatic skeletal muscle injury to investigate the previously unknown role of dendritic cells (DCs) that accumulate in injured tissue. We injected the model antigen ovalbumin (OVA) into the skeletal muscle of injured or sham-treated mice to address the ability of these DCs in antigen uptake, migration, and specific T cell activation in the draining popliteal lymph node (pLN). Immature DC-like cells appeared in the skeletal muscle by 4 days after injury and subsequently acquired a mature phenotype, as indicated by increased expression of the costimulatory molecules CD40 and CD86. After the injection of OVA into the muscle, OVA-loaded DCs migrated into the pLN. The migration of DC-like cells from the injured muscle was enhanced in the presence of the microbial stimulus lipopolysaccharide at the site of antigen uptake and triggered an increased OVA-specific T helper cell type 1 (Th1) response in the pLN. Naïve OVA-loaded DCs were superior in Th1-like priming in the pLN when adoptively transferred into the skeletal muscle of injured mice, a finding indicating the relevance of the microenvironment in the regenerating skeletal muscle for increased Th1-like priming. These findings suggest that DC-like cells that accumulate in the regenerating muscle initiate a protective immune response upon microbial challenge and thereby overcome injury-induced immunosuppression.


Asunto(s)
Inmunidad Adaptativa , Células Dendríticas/citología , Músculo Esquelético/inmunología , Músculo Esquelético/lesiones , Regeneración/fisiología , Traslado Adoptivo , Animales , Antígeno B7-2/metabolismo , Antígenos CD40/metabolismo , Movimiento Celular , Células Dendríticas/inmunología , Endotoxinas , Tolerancia Inmunológica , Células Asesinas Naturales/inmunología , Lipopolisacáridos/metabolismo , Ganglios Linfáticos/inmunología , Activación de Linfocitos , Masculino , Ratones , Ratones Endogámicos BALB C , Ratones Transgénicos , Músculo Esquelético/microbiología , Ovalbúmina , Células TH1/inmunología
17.
J Orthop Trauma ; 30(3): 104-12, 2016 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-26606600

RESUMEN

OBJECTIVE: This review aims to summarize current knowledge regarding the underlying patho-mechanisms of delayed fracture healing in polytraumatized patients. DATA SOURCES AND STUDY SELECTION: The following search terms were used: "fracture", "hemorrhage," "chest trauma," "inflammation," "inflammatory response," "fracture healing," "delayed healing," "nonunion," "fracture stabilisation," "intramedullary nailing," "external fixation," "early total care," and "damage control." Medline, Embase, and Cochrane Library were searched for studies published between January 1, 1990 through March 30, 2014. Of 1322 publications, 68 were included in the current summary. CONCLUSION: Concomitant injuries and the strategy for fracture stabilization seem to affect bone metabolism and fracture healing. Among the relevant patho-mechanisms, interactions between the local and systemic inflammatory response seem to play a role. However, the consequences of fracture fixation strategies in case of severe concomitant injuries on local inflammation and bone healing remain unknown. LEVEL OF EVIDENCE: Prognostic Level IV. See Instructions for Authors for a complete description of levels of evidence.


Asunto(s)
Fijación Interna de Fracturas/estadística & datos numéricos , Curación de Fractura , Fracturas Óseas/epidemiología , Fracturas Óseas/cirugía , Traumatismo Múltiple/epidemiología , Tiempo de Tratamiento/estadística & datos numéricos , Animales , Medicina Basada en la Evidencia , Humanos , Traumatismo Múltiple/terapia , Prevalencia , Medición de Riesgo , Resultado del Tratamiento
18.
PLoS One ; 10(6): e0130155, 2015.
Artículo en Inglés | MEDLINE | ID: mdl-26090673

RESUMEN

Major surgery increases the risk for infectious complications due to the development of immunosuppression. CD56 bright NK cells play a key role in the defense against bacterial infections through the release of Interferon (IFN) γ upon stimulation with monocyte-derived Interleukin (IL) 12. We investigated whether invasive visceral surgery interferes with the IFN-γ synthesis of human NK cells in response to Staphylococcus aureus. In a prospective pilot study, peripheral blood mononuclear cells (PBMC) were isolated from 53 patients before and 1 to 7 d after elective visceral surgery. The release of IL-12 and IFN-γ from PBMC upon exposure to S. aureus in vitro was quantified. The expression of the IL-12 receptor ß1 chain on the surface, the phosphorylation of signal transducer and activator of transcription (STAT) 4, and the synthesis of IFN-γ on/in individual CD56 bright NK cells were investigated using flow cytometry. The modulatory effect of IL-12 on the S. aureus-induced IFN-γ production in CD56 bright NK cells was analyzed. The IFN-γ secretion from purified CD56 bright NK cells was quantified after stimulation with IL-12 and IL-18. After surgery, CD56 bright NK cells among total PBMC were impaired in the release of IFN-γ for at least 5 d. Likewise, the IL-12-induced release of IFN-γ from purified CD56 bright NK cells was abolished. Upon stimulation with S. aureus, PBMC secreted less IL-12 but supplementation with recombinant IL-12 did not restore the capacity of CD56 bright NK cells to produce IFN-γ. CD56 bright NK cells displayed reduced levels of the IL-12Rß1 chain whereas the phosphorylation of STAT4, the key transcription factor for the Ifng gene was not diminished. In summary, after invasive visceral surgery, CD56 bright NK cells are impaired in S. aureus-induced IFN-γ production and might contribute to the enhanced susceptibility to opportunistic infections.


Asunto(s)
Interferón gamma/biosíntesis , Células Asesinas Naturales/inmunología , Células Asesinas Naturales/metabolismo , Infecciones Estafilocócicas/inmunología , Infecciones Estafilocócicas/metabolismo , Staphylococcus aureus/inmunología , Procedimientos Quirúrgicos Operativos/efectos adversos , Anciano , Antígeno CD56/metabolismo , Femenino , Humanos , Inmunomodulación , Inmunofenotipificación , Interleucina-12/metabolismo , Recuento de Leucocitos , Leucocitos Mononucleares/inmunología , Leucocitos Mononucleares/metabolismo , Masculino , Persona de Mediana Edad , Fosforilación , Receptores de Interleucina-12/metabolismo , Factor de Transcripción STAT4/metabolismo , Infecciones Estafilocócicas/microbiología
19.
Clin Sci (Lond) ; 128(11): 825-38, 2015 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-25609031

RESUMEN

Nosocomial infections represent serious complications after traumatic or surgical injuries in intensive care units. The pathogenesis of the underlying immunosuppression is only incompletely understood. In the present study, we investigated whether injury interferes with the function of the adaptive immune system in particular with the differentiation of antigen-specific T helper (Th)-cell responses in vivo. We used a mouse model for traumatic gastrocnemius muscle injury. Ovalbumin (OVA), which served as a foreign model antigen, was injected into the hind footpads for determination of the differentiation of OVA-specific Th-cells in the draining popliteal lymph node (pLN). The release of interferon (IFN)-γ from OVA-specific Th-cells was impaired within 24 h after injury and this impairment persisted for at least 7 days. In contrast, the proliferation of OVA-specific Th-cells remained unaffected. Injury did not modulate the function of antigen-presenting cells (APCs) in the pLN. Adoptive transfer of total T-cells from pLNs of injured mice inhibited IFN-γ production by OVA-specific Th-cells in naive mice. Suppressed Th1 priming did not occur in lymphocyte-deficient mice after injury but was restored by administration of T-cells before injury. Moreover, the suppression of Th1 differentiation required the presence of natural killer (NK) cells that were recruited to the pLN after injury; this recruitment was dependent on lymphocytes, toll-like receptor 4 (TLR4) and myeloid differentiation factor 88 (MyD88). In summary, upon traumatic skeletal muscle injury T-cells and NK cells together prevent the development of protective Th1 immunity. Breaking this co-operation might be a novel approach to reduce the risk of infectious complications after injury.


Asunto(s)
Tolerancia Inmunológica/inmunología , Células Asesinas Naturales/inmunología , Músculo Esquelético/inmunología , Animales , Células Presentadoras de Antígenos/inmunología , Efecto Espectador/inmunología , Diferenciación Celular/inmunología , Células Cultivadas , Citometría de Flujo , Interferón gamma/inmunología , Interferón gamma/metabolismo , Ganglios Linfáticos/citología , Ganglios Linfáticos/inmunología , Masculino , Ratones Endogámicos BALB C , Ratones Noqueados , Músculo Esquelético/lesiones , Factor 88 de Diferenciación Mieloide/genética , Factor 88 de Diferenciación Mieloide/inmunología , Factor 88 de Diferenciación Mieloide/metabolismo , Ovalbúmina/inmunología , Células TH1/inmunología , Células TH1/metabolismo , Receptor Toll-Like 4/genética , Receptor Toll-Like 4/inmunología , Receptor Toll-Like 4/metabolismo
20.
Innate Immun ; 21(2): 115-26, 2015 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-24406749

RESUMEN

Impaired resistance to Pseudomonas aeruginosa-induced pneumonia after cecal ligation and puncture (CLP), a mouse model for human polymicrobial sepsis, is associated with decreased IFN-γ, but increased IL-10, levels in the lung. We investigated the so far unknown mechanisms underlying this reduced IFN-γ synthesis in CLP mice. CD11b(+) NK cells, but not T or NKT cells in the lung were impaired in IFN-γ synthesis upon challenge with Pseudomonas in vitro and in vivo after CLP. The inhibition of NK cells was independent of IL-10. IFN-γ synthesis of NK cells was only partly restored by addition of recombinant IL-12. Accessory cells including dendritic cells and alveolar macrophages were required for maximal IFN-γ secretion. But accessory cells of CLP mice suppressed the IFN-γ secretion from naive lung leukocytes. In turn, naive accessory cells were unable to restore the IFN-γ production from lung leukocytes of CLP mice. Thus, a disturbed interaction of accessory cells and NK cells is involved in the impaired IFN-γ release in response to Pseudomonas in the lung of CLP mice. Considering the importance of IFN-γ in the immune defense against bacteria the dysfunction of accessory cells and NK cells might contribute to the enhanced susceptibility to Pseudomonas after CLP.


Asunto(s)
Células Dendríticas/inmunología , Células Asesinas Naturales/inmunología , Macrófagos Alveolares/inmunología , Infecciones Oportunistas/inmunología , Infecciones por Pseudomonas/inmunología , Pseudomonas aeruginosa/inmunología , Sepsis/inmunología , Animales , Ciego/cirugía , Comunicación Celular , Células Cultivadas , Células Dendríticas/microbiología , Modelos Animales de Enfermedad , Femenino , Humanos , Terapia de Inmunosupresión , Interferón gamma/metabolismo , Células Asesinas Naturales/microbiología , Macrófagos Alveolares/microbiología , Ratones , Ratones Endogámicos BALB C
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA