Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 12 de 12
Filtrar
1.
Blood ; 140(19): 2076-2090, 2022 11 10.
Artigo em Inglês | MEDLINE | ID: mdl-35981499

RESUMO

Graft-versus-host disease (GVHD) remains a major complication after allogeneic hematopoietic stem cell transplantation, a widely used therapy for hematologic malignancies and blood disorders. Here, we report an unexpected role of cytokine leukemia inhibitory factor (LIF) in protecting against GVHD development. Administrating recombinant LIF protein (rLIF) protects mice from GVHD-induced tissue damage and lethality without compromising the graft-versus-leukemia activity, which is crucial to prevent tumor relapse. We found that rLIF decreases the infiltration and activation of donor immune cells and protects intestinal stem cells to ameliorate GVHD. Mechanistically, rLIF downregulates IL-12-p40 expression in recipient dendritic cells after irradiation through activating STAT1 signaling, which results in decreased major histocompatibility complex II levels on intestinal epithelial cells and decreased donor T-cell activation and infiltration. This study reveals a previously unidentified protective role of LIF for GVHD-induced tissue pathology and provides a potential effective therapeutic strategy to limit tissue pathology without compromising antileukemic efficacy.


Assuntos
Doença Enxerto-Hospedeiro , Transplante de Células-Tronco Hematopoéticas , Fator Inibidor de Leucemia , Leucemia , Animais , Camundongos , Doença Enxerto-Hospedeiro/prevenção & controle , Efeito Enxerto vs Leucemia , Transplante de Células-Tronco Hematopoéticas/efeitos adversos , Transplante de Células-Tronco Hematopoéticas/métodos , Leucemia/terapia , Fator Inibidor de Leucemia/genética , Transplante Homólogo
2.
J Immunol ; 204(2): 459-471, 2020 01 15.
Artigo em Inglês | MEDLINE | ID: mdl-31826940

RESUMO

Specific deletion of the tumor suppressor TRAF3 from B lymphocytes in mice leads to the prolonged survival of mature B cells and expanded B cell compartments in secondary lymphoid organs. In the current study, we investigated the metabolic basis of TRAF3-mediated regulation of B cell survival by employing metabolomic, lipidomic, and transcriptomic analyses. We compared the polar metabolites, lipids, and metabolic enzymes of resting splenic B cells purified from young adult B cell-specific Traf3 -/- and littermate control mice. We found that multiple metabolites, lipids, and enzymes regulated by TRAF3 in B cells are clustered in the choline metabolic pathway. Using stable isotope labeling, we demonstrated that phosphocholine and phosphatidylcholine biosynthesis was markedly elevated in Traf3 -/- mouse B cells and decreased in TRAF3-reconstituted human multiple myeloma cells. Furthermore, pharmacological inhibition of choline kinase α, an enzyme that catalyzes phosphocholine synthesis and was strikingly increased in Traf3 -/- B cells, substantially reversed the survival phenotype of Traf3 -/- B cells both in vitro and in vivo. Taken together, our results indicate that enhanced phosphocholine and phosphatidylcholine synthesis supports the prolonged survival of Traf3 -/- B lymphocytes. Our findings suggest that TRAF3-regulated choline metabolism has diagnostic and therapeutic value for B cell malignancies with TRAF3 deletions or relevant mutations.


Assuntos
Linfócitos B/fisiologia , Colina Quinase/metabolismo , Colina/metabolismo , Mieloma Múltiplo/metabolismo , Fator 3 Associado a Receptor de TNF/metabolismo , Animais , Linhagem Celular , Sobrevivência Celular , Colina Quinase/genética , Regulação Neoplásica da Expressão Gênica , Humanos , Camundongos , Camundongos Knockout , Mieloma Múltiplo/genética , Mieloma Múltiplo/patologia , Mutação/genética , Fosforilcolina/metabolismo , Transdução de Sinais , Fator 3 Associado a Receptor de TNF/genética
3.
Front Immunol ; 14: 1167924, 2023.
Artigo em Inglês | MEDLINE | ID: mdl-37207205

RESUMO

Myeloid-derived suppressor cells (MDSCs) are aberrantly expanded in cancer patients and under other pathological conditions. These cells orchestrate the immunosuppressive and inflammatory network to facilitate cancer metastasis and mediate patient resistance to therapies, and thus are recognized as a prime therapeutic target of human cancers. Here we report the identification of the adaptor protein TRAF3 as a novel immune checkpoint that critically restrains MDSC expansion. We found that myeloid cell-specific Traf3-deficient (M-Traf3 -/-) mice exhibited MDSC hyperexpansion during chronic inflammation. Interestingly, MDSC hyperexpansion in M-Traf3 -/- mice led to accelerated growth and metastasis of transplanted tumors associated with an altered phenotype of T cells and NK cells. Using mixed bone marrow chimeras, we demonstrated that TRAF3 inhibited MDSC expansion via both cell-intrinsic and cell-extrinsic mechanisms. Furthermore, we elucidated a GM-CSF-STAT3-TRAF3-PTP1B signaling axis in MDSCs and a novel TLR4-TRAF3-CCL22-CCR4-G-CSF axis acting in inflammatory macrophages and monocytes that coordinately control MDSC expansion during chronic inflammation. Taken together, our findings provide novel insights into the complex regulatory mechanisms of MDSC expansion and open up unique perspectives for the design of new therapeutic strategies that aim to target MDSCs in cancer patients.


Assuntos
Células Supressoras Mieloides , Neoplasias , Fator 3 Associado a Receptor de TNF , Animais , Humanos , Camundongos , Inflamação , Células Mieloides , Neoplasias/metabolismo , Fator 3 Associado a Receptor de TNF/genética , Fator 3 Associado a Receptor de TNF/metabolismo
4.
Front Chem ; 10: 814643, 2022.
Artigo em Inglês | MEDLINE | ID: mdl-35308790

RESUMO

In recent years, the problem of heavy metal pollution has become increasingly prominent, so it is urgent to develop new heavy metal adsorption materials. Compared with many adsorbents, the polyamide-amine dendrimers (PAMAMs) have attracted extensive attention of researchers due to its advantages of macro-molecular cavity, abundant surface functional groups, non-toxicity, high efficiency and easy modification. But in fact, it is not very suitable as an adsorbent because of its solubility and difficulty in separation, which also limits its application in environmental remediation. Therefore, in order to make up for the shortcomings of this material to a certain extent, the synthesis and development of polymer composite materials based on PAMAMs are increasingly prominent in the direction of solving heavy metal pollution. In this paper, the application of composites based on PAMAMs and inorganic or organic components in the adsorption of heavy metal ions is reviewed. Finally, the prospects and challenges of PAMAMs composites for removal of heavy metal ions in water environment are discussed.

5.
ACS Omega ; 7(14): 12231-12241, 2022 Apr 12.
Artigo em Inglês | MEDLINE | ID: mdl-35449935

RESUMO

In this study, the highly crosslinked hyperbranched polyamide-amines (H-PAMAMs) were first prepared via one-pot methods and then modified with thiourea to synthesize a novel adsorbent containing sulfhydryl groups (CHAP-SH), which was used to adsorb Hg(II) ions from aqueous solutions. The adsorption characteristics and mechanism of CHAP-SH for Hg(II) ions were systematically studied. As expected, CHAP-SH exhibited a rapid removal performance toward Hg(II), and the maximum adsorption capacity was 282.74 mg/g at 318 K and pH = 4.5. The whole adsorption behavior could be well described by the pseudo-second-order kinetic model and Langmuir and Redlich-Peterson adsorption isotherm models, which reflected that the adsorption process was mainly monolayer chemisorption. Meanwhile, CHAP-SH had strong selectivity for Hg(II) in the presence of multimetal ions, and it had excellent recoverability after five cycles. In order to further elucidate the adsorption mechanism, the adsorbents before and after adsorption were characterized by Fourier transform infrared spectroscopy, scanning electron microscopy, X-ray photoelectron spectroscopy, thermogravimetric analysis, and energy-dispersive X-ray spectroscopy, and the results showed that the nitrogen-containing, oxygen-containing, and sulfur-containing groups in the adsorbent molecule had synergistic complexation with Hg(II). These results indicated that the adsorbents had great potential in the future treatment of aqueous solutions containing Hg(II).

6.
Cell Stem Cell ; 29(7): 1135-1153.e8, 2022 07 07.
Artigo em Inglês | MEDLINE | ID: mdl-35803230

RESUMO

Microglia are critical in brain development and Alzheimer's disease (AD) etiology. Down syndrome (DS) is the most common genetic developmental disorder and risk factor for AD. Surprisingly, little information is available on the impact of trisomy of human chromosome 21 (Hsa21) on microglial functions during DS brain development and in AD in DS. Using induced pluripotent stem cell (iPSC)-based organoid and chimeric mouse models, we report that DS microglia exhibit an enhanced synaptic pruning function, which alters neuronal synaptic functions. In response to human brain tissue-derived pathological tau, DS microglia undergo cellular senescence and exhibit elevated type-I-interferon signaling. Mechanistically, knockdown of Hsa21-encoded type I interferon receptors, IFNARs, rescues the DS microglial phenotypes both during brain development and in response to pathological tau. Our findings provide in vivo evidence that human microglia respond to pathological tau by exhibiting dystrophic phenotypes. Targeting IFNARs may improve DS microglial functions and prevent senescence.


Assuntos
Doença de Alzheimer , Síndrome de Down , Células-Tronco Pluripotentes Induzidas , Doença de Alzheimer/patologia , Peptídeos beta-Amiloides/metabolismo , Animais , Síndrome de Down/metabolismo , Humanos , Células-Tronco Pluripotentes Induzidas/metabolismo , Interferons/metabolismo , Camundongos , Microglia
7.
Front Oncol ; 11: 737943, 2021.
Artigo em Inglês | MEDLINE | ID: mdl-34778053

RESUMO

The BTK inhibitors ibrutinib and acalabrutinib are FDA-approved drugs for the treatment of B cell malignances. Both drugs have demonstrated clinical efficacy and safety profiles superior to chemoimmunotherapy regimens in patients with chronic lymphocytic leukemia. Mounting preclinical and clinical evidence indicates that both ibrutinib and acalabrutinib are versatile and have direct effects on many immune cell subsets as well as other cell types beyond B cells. The versatility and immunomodulatory effects of both drugs have been exploited to expand their therapeutic potential in a wide variety of human diseases. Over 470 clinical trials are currently registered at ClinicalTrials.gov to test the efficacy of ibrutinib or acalabrutinib not only in almost every type of B cell malignancies, but also in hematological malignancies of myeloid cells and T cells, solid tumors, chronic graft versus host disease (cGHVD), autoimmune diseases, allergy and COVID-19 (http:www.clinicaltrials.gov). In this review, we present brief discussions of the clinical trials and relevant key preclinical evidence of ibrutinib and acalabrutinib as monotherapies or as part of combination therapies for the treatment of human diseases beyond B cell malignancies. Adding to the proven efficacy of ibrutinib for cGVHD, preliminary results of clinical trials have shown promising efficacy of ibrutinib or acalabrutinib for certain T cell malignancies, allergies and severe COVID-19. However, both BTK inhibitors have no or limited efficacy for refractory or recurrent solid tumors. These clinical data together with additional pending results from ongoing trials will provide valuable information to guide the design and improvement of future trials, including optimization of combination regimens and dosing sequences as well as better patient stratification and more efficient delivery strategies. Such information will further advance the precise implementation of BTK inhibitors into the clinical toolbox for the treatment of different human diseases.

8.
Front Cell Dev Biol ; 9: 727531, 2021.
Artigo em Inglês | MEDLINE | ID: mdl-34485307

RESUMO

The clinical success of the two BTK inhibitors, ibrutinib and acalabrutinib, represents a major breakthrough in the treatment of chronic lymphocytic leukemia (CLL) and has also revolutionized the treatment options for other B cell malignancies. Increasing evidence indicates that in addition to their direct effects on B lymphocytes, both BTK inhibitors also directly impact the homeostasis, phenotype and function of many other cell subsets of the immune system, which contribute to their high efficacy as well as adverse effects observed in CLL patients. In this review, we attempt to provide an overview on the overlapping and differential effects of ibrutinib and acalabrutinib on specific receptor signaling pathways in different immune cell subsets other than B cells, including T cells, NK cells, monocytes, macrophages, granulocytes, myeloid-derived suppressor cells, dendritic cells, osteoclasts, mast cells and platelets. The shared and distinct effects of ibrutinib versus acalabrutinib are mediated through BTK-dependent and BTK-independent mechanisms, respectively. Such immunomodulatory effects of the two drugs have fueled myriad explorations of their repurposing opportunities for the treatment of a wide variety of other human diseases involving immune dysregulation.

9.
Front Immunol ; 12: 670338, 2021.
Artigo em Inglês | MEDLINE | ID: mdl-34745083

RESUMO

Proteins controlling mitochondrial fission have been recognized as essential regulators of mitochondrial functions, mitochondrial quality control and cell apoptosis. In the present study, we identified the critical B cell survival regulator TRAF3 as a novel binding partner of the key mitochondrial fission factor, MFF, in B lymphocytes. Elicited by our unexpected finding that the majority of cytoplasmic TRAF3 proteins were localized at the mitochondria in resting splenic B cells after ex vivo culture for 2 days, we found that TRAF3 specifically interacted with MFF as demonstrated by co-immunoprecipitation and GST pull-down assays. We further found that in the absence of stimulation, increased protein levels of mitochondrial TRAF3 were associated with altered mitochondrial morphology, decreased mitochondrial respiration, increased mitochondrial ROS production and membrane permeabilization, which eventually culminated in mitochondria-dependent apoptosis in resting B cells. Loss of TRAF3 had the opposite effects on the morphology and function of mitochondria as well as mitochondria-dependent apoptosis in resting B cells. Interestingly, co-expression of TRAF3 and MFF resulted in decreased phosphorylation and ubiquitination of MFF as well as decreased ubiquitination of TRAF3. Moreover, lentivirus-mediated overexpression of MFF restored mitochondria-dependent apoptosis in TRAF3-deficient malignant B cells. Taken together, our findings provide novel insights into the apoptosis-inducing mechanisms of TRAF3 in B cells: as a result of survival factor deprivation or under other types of stress, TRAF3 is mobilized to the mitochondria through its interaction with MFF, where it triggers mitochondria-dependent apoptosis. This new role of TRAF3 in controlling mitochondrial homeostasis might have key implications in TRAF3-mediated regulation of B cell transformation in different cellular contexts. Our findings also suggest that mitochondrial fission is an actionable therapeutic target in human B cell malignancies, including those with TRAF3 deletion or relevant mutations.


Assuntos
Linfócitos B/fisiologia , Dinâmica Mitocondrial/fisiologia , Fator 3 Associado a Receptor de TNF/fisiologia , Animais , Apoptose , Linhagem Celular Tumoral , Respiração Celular , Sobrevivência Celular , Dinaminas/genética , Humanos , Proteínas de Membrana/genética , Proteínas de Membrana/fisiologia , Camundongos , Proteínas Mitocondriais/genética , Proteínas Mitocondriais/fisiologia , Espécies Reativas de Oxigênio/metabolismo , Fator 3 Associado a Receptor de TNF/análise
10.
J Vis Exp ; (139)2018 09 07.
Artigo em Inglês | MEDLINE | ID: mdl-30247482

RESUMO

Antibodies, also termed as immunoglobulins (Ig), secreted by differentiated B lymphocytes, plasmablasts/plasma cells, in humoral immunity provide a formidable defense against invading pathogens via diverse mechanisms. One major goal of vaccination is to induce protective antigen-specific antibodies to prevent life-threatening infections. Both thymus-dependent (TD) and thymus-independent (TI) antigens can elicit robust antigen-specific IgM responses and can also induce the production of isotype-switched antibodies (IgG, IgA and IgE) as well as the generation of memory B cells with the help provided by antigen presenting cells (APCs). Here, we describe a protocol to characterize TD and TI Ig isotype responses in mice using enzyme-linked immunosorbent assay (ELISA). In this protocol, TD and TI Ig responses are elicited in mice by intraperitoneal (i.p.) immunization with hapten-conjugated model antigens TNP-KLH (in alum) and TNP-polysaccharide (in PBS), respectively. To induce TD memory response, a booster immunization of TNP-KLH in alum is given at 3 weeks after the first immunization with the same antigen/adjuvant. Mouse sera are harvested at different time points before and after immunization. Total serum Ig levels and TNP-specific antibodies are subsequently quantified using Ig isotype-specific Sandwich and indirect ELISA, respectively. In order to correctly quantify the serum concentration of each Ig isotype, the samples need to be appropriately diluted to fit within the linear range of the standard curves. Using this protocol, we have consistently obtained reliable results with high specificity and sensitivity. When used in combination with other complementary methods such as flow cytometry, in vitro culture of splenic B cells and immunohistochemical staining (IHC), this protocol will allow researchers to gain a comprehensive understanding of antibody responses in a given experimental setting.


Assuntos
Formação de Anticorpos/imunologia , Antígenos T-Independentes/imunologia , Ensaio de Imunoadsorção Enzimática/métodos , Isotipos de Imunoglobulinas/imunologia , Linfócitos T/imunologia , Animais , Camundongos
11.
Curr Pharmacol Rep ; 4(1): 64-90, 2018 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-29527458

RESUMO

PURPOSE OF REVIEW: This review presents an overview of the current knowledge of TRAF molecules in inflammation with an emphasis on available human evidence and direct in vivo evidence of mouse models that demonstrate the contribution of TRAF molecules in the pathogenesis of inflammatory diseases. RECENT FINDINGS: The tumor necrosis factor receptor (TNF-R)-associated factor (TRAF) family of cytoplasmic proteins was initially identified as signaling adaptors that bind directly to the intracellular domains of receptors of the TNF-R superfamily. It is now appreciated that TRAF molecules are widely employed in signaling by a variety of adaptive and innate immune receptors as well as cytokine receptors. TRAF-dependent signaling pathways typically lead to the activation of nuclear factor-κBs (NF-κBs), mitogen-activated protein kinases (MAPKs), or interferon-regulatory factors (IRFs). Most of these signaling pathways have been linked to inflammation, and therefore TRAF molecules were expected to regulate inflammation and inflammatory responses since their discovery in 1990s. However, direct in vivo evidence of TRAFs in inflammation and especially in inflammatory diseases had been lacking for many years, partly due to the difficulty imposed by early lethality of TRAF2-/-, TRAF3-/-, and TRAF6-/- mice. With the creation of conditional knockout and lineage-specific transgenic mice of different TRAF molecules, our understanding about TRAFs in inflammation and inflammatory responses has rapidly advanced during the past decade. SUMMARY: Increasing evidence indicates that TRAF molecules are versatile and indispensable regulators of inflammation and inflammatory responses and that aberrant expression or function of TRAFs contributes to the pathogenesis of inflammatory diseases.

12.
Front Immunol ; 9: 2111, 2018.
Artigo em Inglês | MEDLINE | ID: mdl-30294322

RESUMO

The tumor necrosis factor receptor (TNF-R)-associated factor (TRAF) family of cytoplasmic adaptor proteins regulate the signal transduction pathways of a variety of receptors, including the TNF-R superfamily, Toll-like receptors (TLRs), NOD-like receptors (NLRs), RIG-I-like receptors (RLRs), and cytokine receptors. TRAF-dependent signaling pathways participate in a diverse array of important cellular processes, including the survival, proliferation, differentiation, and activation of different cell types. Many of these TRAF-dependent signaling pathways have been implicated in cancer pathogenesis. Here we analyze the current evidence of genetic alterations of TRAF molecules available from The Cancer Genome Atlas (TCGA) and the Catalog of Somatic Mutations in Cancer (COSMIC) as well as the published literature, including copy number variations and mutation landscape of TRAFs in various human cancers. Such analyses reveal that both gain- and loss-of-function genetic alterations of different TRAF proteins are commonly present in a number of human cancers. These include pancreatic cancer, meningioma, breast cancer, prostate cancer, lung cancer, liver cancer, head and neck cancer, stomach cancer, colon cancer, bladder cancer, uterine cancer, melanoma, sarcoma, and B cell malignancies, among others. Furthermore, we summarize the key in vivo and in vitro evidence that demonstrates the causal roles of genetic alterations of TRAF proteins in tumorigenesis within different cell types and organs. Taken together, the information presented in this review provides a rationale for the development of therapeutic strategies to manipulate TRAF proteins or TRAF-dependent signaling pathways in different human cancers by precision medicine.


Assuntos
Carcinogênese/genética , Neoplasias/genética , Transdução de Sinais/genética , Peptídeos e Proteínas Associados a Receptores de Fatores de Necrose Tumoral/genética , Diferenciação Celular/genética , Proliferação de Células/genética , Sobrevivência Celular/genética , Variações do Número de Cópias de DNA , Humanos , Mutação , Neoplasias/tratamento farmacológico , Neoplasias/patologia , Medicina de Precisão/métodos , Peptídeos e Proteínas Associados a Receptores de Fatores de Necrose Tumoral/metabolismo
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA