Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 10 de 10
Filter
Add more filters










Publication year range
1.
Trends Mol Med ; 30(8): 699-701, 2024 Aug.
Article in English | MEDLINE | ID: mdl-39048417

Subject(s)
Science , Humans
2.
Trends Immunol ; 45(7): 486-494, 2024 Jul.
Article in English | MEDLINE | ID: mdl-38876831

ABSTRACT

Immunotherapies have revolutionized the treatment of certain cancers, but challenges remain in overcoming immunotherapy resistance. Research shows that metabolic modulation of the tumor microenvironment can enhance antitumor immunity. Here, we discuss recent preclinical and clinical evidence for the efficacy of combining metabolic modifiers with immunotherapies. While this combination holds great promise, a few key areas must be addressed, which include identifying the effects of metabolic modifiers on immune cell metabolism, the putative biomarkers of therapeutic efficacy, the efficacy of modifiers on tumors harboring metabolic heterogeneity, and the potential development of resistance due to tumor reliance on alternative metabolic pathways. We propose solutions to these problems and posit that assessing these parameters is crucial for considering the potential of metabolic modifiers in sensitizing tumors to immunotherapies.


Subject(s)
Drug Resistance, Neoplasm , Immunotherapy , Metabolic Networks and Pathways , Neoplasms , Tumor Microenvironment , Humans , Neoplasms/immunology , Neoplasms/therapy , Neoplasms/metabolism , Immunotherapy/methods , Tumor Microenvironment/immunology , Animals , Drug Resistance, Neoplasm/immunology
3.
Trends Mol Med ; 30(8): 765-780, 2024 Aug.
Article in English | MEDLINE | ID: mdl-38821771

ABSTRACT

While immunomodulatory imide drugs (IMiDs) have been authorised for treatment of haematological cancers for over two decades, the appreciation of their ability to stimulate antitumour T cell and natural killer (NK) cell responses is relatively recent. Clinical trial data increasingly show that targeted immunotherapies, such as antibodies, T cells, and vaccines, improve outcomes when delivered in combination with the IMiD derivatives lenalidomide or pomalidomide. Here, we review these clinical data to highlight the relevance of IMiDs in combinatorial immunotherapy for both haematological and solid tumours. Further research into the molecular mechanisms of IMiDs and an increased understanding of their immunomodulatory effects may refine the specific applications of IMiDs and improve the design of future clinical trials, moving IMiDs to the forefront of combinatorial cancer immunotherapy.


Subject(s)
Immunomodulating Agents , Immunotherapy , Neoplasms , Humans , Immunotherapy/methods , Neoplasms/immunology , Neoplasms/drug therapy , Neoplasms/therapy , Immunomodulating Agents/therapeutic use , Immunomodulating Agents/pharmacology , Animals , Thalidomide/analogs & derivatives , Thalidomide/therapeutic use , Thalidomide/pharmacology , Immunomodulation/drug effects , Clinical Trials as Topic
4.
Cancer Lett ; 520: 295-306, 2021 11 01.
Article in English | MEDLINE | ID: mdl-34389433

ABSTRACT

Interleukin (IL-6) is a pleotropic cytokine with both tumor-promoting and -inhibitory effects on breast cancer growth. However, the mechanisms governing the outcome of IL-6 on cancer progression remain to be clarified. Our study unraveled a novel long noncoding RNA (lncRNA) AU021063 downstream of IL-6 signaling. We found that IL-6 induced the expression of AU021063 predominantly in breast cancer compared to other cancer types. Mechanistically, IL-6 induced AT-rich interactive domain 5a (Arid5a) expression, which promotes the transcription of AU021063. In turn, AU021063 promotes breast cancer metastasis through stabilizing tribbles homolog 3 (Trib3) and activating Mek/Erk signaling pathway. Genetic ablation of either Arid5a, AU021063 or Trib3 abolished breast cancer metastasis in vitro and in vivo. Overall, our study highlights the importance of IL-6-Arid5a-AU021063 axis in regulating breast cancer invasiveness and metastasis, which may provide potential novel therapeutics for breast cancer.


Subject(s)
Breast Neoplasms/genetics , Cell Cycle Proteins/genetics , DNA-Binding Proteins/genetics , Interleukin-6/genetics , RNA, Long Noncoding/genetics , Transcription Factors/genetics , Animals , Breast Neoplasms/pathology , Cell Line, Tumor , Cell Movement/genetics , Cell Proliferation/genetics , Disease Models, Animal , Female , Humans , MAP Kinase Signaling System/genetics , Mice , Neoplasm Invasiveness/genetics , Neoplasm Invasiveness/pathology , Neoplasm Metastasis , Signal Transduction
5.
Sci Signal ; 13(624)2020 03 24.
Article in English | MEDLINE | ID: mdl-32209697

ABSTRACT

The lipopolysaccharide (LPS)-induced endocytosis of Toll-like receptor 4 (TLR4) is an essential step in the production of interferon-ß (IFN-ß), which activates the transcription of antiviral response genes by STAT1 phosphorylated at Tyr701 Here, we showed that STAT1 regulated proinflammatory cytokine production downstream of TLR4 endocytosis independently of IFN-ß signaling and the key proinflammatory regulator NF-κB. In human macrophages, TLR4 endocytosis activated a noncanonical phosphorylation of STAT1 at Thr749, which subsequently promoted the production of interleukin-6 (IL-6) and IL-12p40 through distinct mechanisms. STAT1 phosphorylated at Thr749 activated the expression of the gene encoding ARID5A, which stabilizes IL6 mRNA. Moreover, STAT1 phosphorylated at Thr749 directly enhanced transcription of the gene encoding IL-12p40 (IL12B). Instead of affecting STAT1 nuclear translocation, phosphorylation of Thr749 facilitated the binding of STAT1 to a noncanonical DNA motif (5'-TTTGANNC-3') in the promoter regions of ARID5A and IL12B The endocytosis of TLR4 induced the formation of a complex between the kinases TBK1 and IKKß, which mediated the phosphorylation of STAT1 at Thr749 Our data suggest that noncanonical phosphorylation in response to LPS confers STAT1 with distinct DNA binding and gene-regulatory properties that promote both IL12B expression and IL6 mRNA stabilization. Thus, our study provides a potential mechanism for how TLR4 endocytosis might regulate proinflammatory cytokine production.


Subject(s)
Interleukin-12 Subunit p40/biosynthesis , Interleukin-6/biosynthesis , Lipopolysaccharides/pharmacology , STAT1 Transcription Factor/metabolism , Transcription, Genetic/drug effects , HEK293 Cells , Humans , Interleukin-12 Subunit p40/genetics , Interleukin-6/genetics , Phosphorylation/drug effects , Phosphorylation/genetics , STAT1 Transcription Factor/genetics , THP-1 Cells
6.
Proc Natl Acad Sci U S A ; 116(30): 15128-15133, 2019 07 23.
Article in English | MEDLINE | ID: mdl-31289228

ABSTRACT

Immune cells infiltrate adipose tissues and provide a framework to regulate energy homeostasis. However, the precise underlying mechanisms and signaling by which the immune system regulates energy homeostasis in metabolic tissues remain poorly understood. Here, we show that the AT-rich interactive domain 5A (Arid5a), a cytokine-induced nucleic acid binding protein, is important for the maintenance of adipose tissue homeostasis. Long-term deficiency of Arid5a in mice results in adult-onset severe obesity. In contrast, transgenic mice overexpressing Arid5a are highly resistant to high-fat diet-induced obesity. Inhibition of Arid5a facilitates the in vitro differentiation of 3T3-L1 cells and fibroblasts to adipocytes, whereas its induction substantially inhibits their differentiation. Molecular studies reveal that Arid5a represses the transcription of peroxisome proliferator activated receptor gamma 2 (Ppar-γ2) due to which, in the absence of Arid5a, Ppar-γ2 is persistently expressed in fibroblasts. This phenomenon is accompanied by enhanced fatty acid uptake in Arid5a-deficient cells, which shifts metabolic homeostasis toward prolipid metabolism. Furthermore, we show that Arid5a and Ppar-γ2 are dynamically counterregulated by each other, hence maintaining adipogenic homeostasis. Thus, we show that Arid5a is an important negative regulator of energy metabolism and can be a potential target for metabolic disorders.


Subject(s)
Adipogenesis/genetics , Adipose Tissue/metabolism , DNA-Binding Proteins/genetics , Feedback, Physiological , Obesity/genetics , PPAR gamma/genetics , Transcription Factors/genetics , 3T3-L1 Cells , Adipocytes/metabolism , Adipocytes/pathology , Adipose Tissue/pathology , Animals , Biological Transport , Cell Differentiation , DNA-Binding Proteins/metabolism , Diet, High-Fat/adverse effects , Energy Metabolism/genetics , Fatty Acids/metabolism , Female , Gene Expression Regulation , Homeostasis/genetics , Male , Mice , Mice, Inbred C57BL , Mice, Transgenic , Obesity/etiology , Obesity/metabolism , Obesity/pathology , PPAR gamma/metabolism , Signal Transduction , Transcription Factors/metabolism
7.
Proc Natl Acad Sci U S A ; 115(46): 11802-11807, 2018 11 13.
Article in English | MEDLINE | ID: mdl-30373817

ABSTRACT

Immunomodulatory drugs (IMiDs), including thalidomide derivatives such as lenalidomide and pomalidomide, offer therapeutic benefit in several hematopoietic malignancies and autoimmune/inflammatory diseases. However, it is difficult to study the IMiD mechanism of action in murine disease models because murine cereblon (CRBN), the substrate receptor for IMiD action, is resistant to some of IMiDs therapeutic effects. To overcome this difficulty, we generated humanized cereblon (CRBNI391V) mice thereby providing an animal model to unravel complex mechanisms of action in a murine physiological setup. In our current study, we investigated the degradative effect toward IKZF1 and CK-1α, a target substrate of IMiDs. Unlike WT mice which were resistant to lenalidomide and pomalidomide, T lymphocytes from CRBNI391V mice responded with a higher degree of IKZF1 and CK-1α protein degradation. Furthermore, IMiDs resulted in an increase in IL-2 among CRBNI391V mice but not in the WT group. We have also tested a thalidomide derivative, FPFT-2216, which showed an inhibitory effect toward IKZF1 protein level. As opposed to pomalidomide, FPFT-2216 and lenalidomide degrades CK-1α. Additionally, we assessed the potential therapeutic effects of IMiDs in dextran sodium sulfate (DSS)-induced colitis. In both WT and humanized mice, lenalidomide showed a significant therapeutic effect in the DSS model of colitis, while the effect of pomalidomide was less pronounced. Thus, while IMiDs' degradative effect on IKZF1 and CK-1α, and up-regulation of IL-2, is dependent on CRBN, the therapeutic benefit of IMiDs in a mouse model of inflammatory bowel disease occurs through a CRBN-IMiD binding region independent pathway.


Subject(s)
Immunomodulation/drug effects , Immunomodulation/physiology , Nerve Tissue Proteins/drug effects , Adaptor Proteins, Signal Transducing , Animals , Humans , Ikaros Transcription Factor/drug effects , Ikaros Transcription Factor/metabolism , Immunologic Factors/metabolism , Mice , Models, Animal , Nerve Tissue Proteins/genetics , Nerve Tissue Proteins/metabolism , Nerve Tissue Proteins/physiology , Peptide Hydrolases/genetics , Peptide Hydrolases/metabolism , Proteolysis/drug effects , Substrate Specificity , Ubiquitin-Protein Ligases/metabolism
8.
Proc Natl Acad Sci U S A ; 113(38): 10625-30, 2016 09 20.
Article in English | MEDLINE | ID: mdl-27601648

ABSTRACT

Immunomodulatory drugs (IMiDs) are a family of compounds derived from thalidomide. Binding of the IMiD molecule to the Lon protease Cereblon initiates the degradation of substrates via the ubiquitin proteasome pathway. Here, we show that Cereblon forms a complex with Rabex-5, a regulator of immune homeostasis. Treatment with lenalidomide prevented the association of Cereblon with Rabex-5. Conversely, mutation of the IMiD binding site increased Cereblon-Rabex-5 coimmunoprecipitation. The thalidomide binding region of Cereblon therefore regulates the formation of this complex. Knockdown of Rabex-5 in the THP-1 macrophage cell line up-regulated Toll-like receptor (TLR)-induced cytokine and type 1 IFN production via a STAT1/IRF activating pathway. Thus, we identify Rabex-5 as a IMiD target molecule that functions to restrain TLR activated auto-immune promoting pathways. We propose that release of Rabex-5 from complex with Cereblon enables the suppression of immune responses, contributing to the antiinflammatory properties of IMiDs.


Subject(s)
Guanine Nucleotide Exchange Factors/metabolism , Immunity, Cellular/genetics , Interferon Type I/genetics , Peptide Hydrolases/metabolism , Adaptor Proteins, Signal Transducing , Binding Sites/drug effects , Gene Knockdown Techniques , Guanine Nucleotide Exchange Factors/genetics , Humans , Immunity, Cellular/drug effects , Immunologic Factors/pharmacology , Interferon Type I/biosynthesis , Lenalidomide , Multiprotein Complexes/drug effects , Multiprotein Complexes/genetics , Peptide Hydrolases/genetics , Protein Binding/drug effects , Thalidomide/analogs & derivatives , Thalidomide/pharmacology , Toll-Like Receptors/genetics , Toll-Like Receptors/metabolism , Ubiquitin-Protein Ligases
9.
Int Breastfeed J ; 11: 4, 2016.
Article in English | MEDLINE | ID: mdl-26997971

ABSTRACT

BACKGROUND: Exclusive breastfeeding for the first 6 months of life is recommended internationally. This study aimed to investigate exclusive breastfeeding practices and associated factors among mothers of infants aged less than 6 months. METHODS: A community based cross-sectional study was conducted on mother-infant pairs in Dubti town in May, 2015. In this study, exclusive breastfeeding was defined as an infant's breast milk consumption without supplementation of any type of food or drink, except for vitamins, minerals and necessary medications in the 24 h preceding the survey. Descriptive statistics, bivariate and multivariable logistic regression analysis were employed to identify the factors associated with exclusive breastfeeding practices. Variables with a p-value < 0.05 in the multivariable model were identified as predictors of exclusive breastfeeding practices. RESULTS: Exclusive breastfeeding under 6 months was practiced by 81.1 % (95 % Confidence Interval [CI] 77.0, 85.0 %) of mothers of infants aged less than 6 months. The median duration of exclusive breastfeeding for infants less than 6 months was 3 months. Multivariable logistic regression analysis showed that initiation of breastfeeding within 1 h after birth (Adjusted Odds Ratio [AOR] 5.46; 95 % CI 1.93, 15.41), age of infants of less than 2 months (AOR 7.03; 95 % CI 2.16, 22.88), being a housewife (AOR 4.81; 95 % CI 2.30, 10.06) and mothers who received postnatal counseling (AOR 3.88; 95 % CI 1.88, 7.99) were positive predictors of exclusive breastfeeding. CONCLUSION: The study revealed that exclusive breastfeeding under 6 months using 24-h recall method was lower than the World Health Organization recommendation. Therefore, interventions could focus on educating mothers the importance of timely initiation of breastfeeding and postnatal care in the study area.

10.
Int Immunol ; 28(6): 307-15, 2016 06.
Article in English | MEDLINE | ID: mdl-26865412

ABSTRACT

Thalidomide and its derivatives, collectively referred to as immunomodulatory drugs (IMiDs), are effective inhibitors of inflammation and are known to inhibit TLR-induced TNFα production. The identification of Cereblon as the receptor for these compounds has led to a rapid advancement in our understanding of IMiD properties; however, there remain no studies addressing the role of Cereblon in mediating the suppressive effect of IMiDs on TLR responses. Here, we developed Cereblon-deficient mice using the CRISPR-Cas9 system. TLR-induced cytokine responses were unaffected by Cereblon deficiency in vivo Moreover, IMiD treatment inhibited cytokine production even in the absence of Cereblon. The IMiD-induced suppression of cytokine production therefore occurs independently of Cereblon in mice. Further investigation revealed that IMiDs are potent inhibitors of TLR-induced type-1 interferon production via suppression of the TRIF/IRF3 pathway. These data suggest that IMiDs may prove effective in the treatment of disorders characterized by the ectopic production of type-1 interferon. Significantly, these properties are mediated separately from thalidomide's teratogenic receptor, Cereblon. Thus, certain therapeutic properties of Thalidomide can be separated from its harmful side effects.


Subject(s)
Immunologic Factors/therapeutic use , Inflammation/drug therapy , Macrophages/drug effects , Nerve Tissue Proteins/metabolism , Thalidomide/analogs & derivatives , Thalidomide/therapeutic use , Adaptor Proteins, Signal Transducing , Adaptor Proteins, Vesicular Transport/metabolism , Animals , Cells, Cultured , Clustered Regularly Interspaced Short Palindromic Repeats , Immunosuppression Therapy , Interferon Regulatory Factor-3/metabolism , Interferon Type I/metabolism , Lenalidomide , Macrophages/immunology , Mice , Mice, Inbred C57BL , Mice, Knockout , Nerve Tissue Proteins/genetics , Signal Transduction/drug effects , Toll-Like Receptor 4/metabolism , Tumor Necrosis Factor-alpha/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL