Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 24
Filter
Add more filters










Publication year range
1.
Nat Chem Biol ; 2024 Mar 20.
Article in English | MEDLINE | ID: mdl-38509349

ABSTRACT

Angiogenic programming in the vascular endothelium is a tightly regulated process for maintaining tissue homeostasis and is activated in tissue injury and the tumor microenvironment. The metabolic basis of how gas signaling molecules regulate angiogenesis is elusive. Here, we report that hypoxic upregulation of ·NO in endothelial cells reprograms the transsulfuration pathway to increase biogenesis of hydrogen sulfide (H2S), a proangiogenic metabolite. However, decreased H2S oxidation due to sulfide quinone oxidoreductase (SQOR) deficiency synergizes with hypoxia, inducing a reductive shift and limiting endothelial proliferation that is attenuated by dissipation of the mitochondrial NADH pool. Tumor xenografts in whole-body (WBCreSqorfl/fl) and endothelial-specific (VE-cadherinCre-ERT2Sqorfl/fl) Sqor-knockout mice exhibit lower mass and angiogenesis than control mice. WBCreSqorfl/fl mice also exhibit decreased muscle angiogenesis following femoral artery ligation compared to control mice. Collectively, our data reveal the molecular intersections between H2S, O2 and ·NO metabolism and identify SQOR inhibition as a metabolic vulnerability for endothelial cell proliferation and neovascularization.

2.
Am J Physiol Gastrointest Liver Physiol ; 326(1): G53-G66, 2024 01 01.
Article in English | MEDLINE | ID: mdl-37933447

ABSTRACT

Neutrophils are abundant immune cells in the colon tumor microenvironment. Studies have shown that neutrophils are recruited into hypoxic foci in colon cancer. However, the impact of hypoxia signaling on neutrophil function and its involvement in colon tumorigenesis remain unclear. To address this, we generated mice with a deletion of hypoxia-inducible factor (HIF)-1α or HIF-2α in neutrophils driven by the MRP8Cre (HIF-1αΔNeu) or (HIF-2αΔNeu) and littermate controls. In an azoxymethane (AOM)/dextran sulfate sodium (DSS) model of colon cancer, the disruption of neutrophils-HIF-1α did not result in any significant changes in body weight, colon length, tumor size, proliferation, or burden. However, the disruption of HIF-2α in neutrophils led to a slight increase in body weight, a significant decrease in the number of tumors, and a reduction in tumor size and volume compared with their littermate controls. Histological analysis of colon tissue from mice with HIF-2α-deficient neutrophils revealed notable reductions in proliferation as compared with control mice. In addition, we observed reduced levels of proinflammatory cytokines, such as TNF-α and IL-1ß, in neutrophil-specific HIF-2α-deficient mice in both the tumor tissue as well as the neutrophils. Importantly, it is worth noting that the reduced tumorigenesis associated with HIF-2α deficiency in neutrophils was not evident in already established syngeneic tumors or a DSS-induced inflammation model, indicating a potential role of HIF-2α specifically in colon tumorigenesis. In conclusion, we found that the loss of neutrophil-specific HIF-2α slows colon tumor growth and progression by reducing the levels of inflammatory mediators.NEW & NOTEWORTHY Despite the importance of hypoxia and neutrophils in colorectal cancer (CRC), the contribution of neutrophil-specific HIFs to colon tumorigenesis is not known. We describe that neutrophil HIF-1α has no impact on colon cancer, whereas neutrophil HIF-2α loss reduces CRC growth by decreasing proinflammatory and immunosuppressive cytokines. Furthermore, neutrophil HIF-2α does not reduce preestablished tumor growth or inflammation-induced colitis. The present study offers novel potential of neutrophil HIF-2α as a therapeutic target in CRC.


Subject(s)
Colitis-Associated Neoplasms , Colonic Neoplasms , Animals , Mice , Basic Helix-Loop-Helix Transcription Factors/genetics , Body Weight , Carcinogenesis/pathology , Cell Transformation, Neoplastic/pathology , Colitis-Associated Neoplasms/genetics , Colonic Neoplasms/genetics , Colonic Neoplasms/pathology , Cytokines , Hypoxia , Hypoxia-Inducible Factor 1, alpha Subunit/genetics , Inflammation , Neutrophils , Tumor Microenvironment
3.
Exp Hematol ; 125-126: 25-36.e1, 2023.
Article in English | MEDLINE | ID: mdl-37562670

ABSTRACT

Dietary consumption serves as the primary source of iron uptake, and erythropoiesis acts as a major regulator of systemic iron demand. In addition to intestinal iron absorption, macrophages play a crucial role in recycling iron from senescent red blood cells. The kidneys are responsible for the production of erythropoietin (Epo), which stimulates erythropoiesis, whereas the liver plays a central role in producing the iron-regulatory hormone hepcidin. The transcriptional regulator hypoxia-inducible factor (HIF)2α has a central role in the regulation of Epo, hepcidin, and intestinal iron absorption and therefore plays a crucial role in coordinating the tissue crosstalk to maintain systemic iron demands. However, the precise involvement of Hif2α in macrophages in terms of iron homeostasis remains uncertain. Our study demonstrates that deleting Hif2α in macrophages does not disrupt the expression of iron transporters or basal erythropoiesis. Mice lacking Hif2α in myeloid cells exhibited no discernible differences in hemodynamic parameters, including hemoglobin concentrations and erythrocyte count, when compared with littermate controls. This similarity was observed under conditions of both dietary iron deficiency and acute erythropoietic demand. Notably, we observed a significant increase in the expression of iron transporters in the duodenum during iron deficiency, indicating heightened iron absorption. Therefore, our findings suggest that the disruption of Hif2α in myeloid cells does not significantly impact systemic iron homeostasis under normal physiologic conditions. However, its disruption induces adaptive physiologic changes in response to elevated iron demand, potentially serving as a mechanism to sustain increased erythropoietic demand.


Subject(s)
Erythropoietin , Iron Deficiencies , Animals , Mice , Erythropoiesis , Erythropoietin/genetics , Erythropoietin/metabolism , Hepcidins/genetics , Homeostasis , Iron/metabolism
4.
J Biol Chem ; 299(5): 104691, 2023 05.
Article in English | MEDLINE | ID: mdl-37037306

ABSTRACT

Mitophagy is a cargo-specific autophagic process that recycles damaged mitochondria to promote mitochondrial turnover. PTEN-induced putative kinase 1 (PINK1) mediates the canonical mitophagic pathway. However, the role of PINK1 in diseases where mitophagy has been purported to play a role, such as colorectal cancer, is unclear. Our results here demonstrate that higher PINK1 expression is positively correlated with decreased colon cancer survival, and mitophagy is required for colon cancer growth. We show that doxycycline-inducible knockdown (KD) of PINK1 in a panel of colon cancer cell lines inhibited proliferation, whereas disruption of other mitophagy receptors did not impact cell growth. We observed that PINK KD led to a decrease in mitochondrial respiration, membrane hyperpolarization, accumulation of mitochondrial DNA, and depletion of antioxidant glutathione. In addition, mitochondria are important hubs for the utilization of iron and synthesizing iron-dependent cofactors such as heme and iron sulfur clusters. We observed an increase in the iron storage protein ferritin and a decreased labile iron pool in the PINK1 KD cells, but total cellular iron or markers of iron starvation/overload were not affected. Finally, cellular iron storage and the labile iron pool are maintained via autophagic degradation of ferritin (ferritinophagy). We found overexpressing nuclear receptor coactivator 4, a key adaptor for ferritinophagy, rescued cell growth and the labile iron pool in PINK1 KD cells. These results indicate that PINK1 integrates mitophagy and ferritinophagy to regulate intracellular iron availability and is essential for maintaining intracellular iron homeostasis to support survival and growth in colorectal cancer cells.


Subject(s)
Colonic Neoplasms , Colorectal Neoplasms , Mitophagy , Protein Kinases , Humans , Colonic Neoplasms/genetics , Colonic Neoplasms/pathology , Colorectal Neoplasms/genetics , Colorectal Neoplasms/pathology , Ferritins , Iron/metabolism , Protein Kinases/genetics , Protein Kinases/metabolism , Ubiquitin-Protein Ligases/metabolism
5.
bioRxiv ; 2023 Mar 15.
Article in English | MEDLINE | ID: mdl-36993187

ABSTRACT

Angiogenic programming in the vascular endothelium is a tightly regulated process to maintain tissue homeostasis and is activated in tissue injury and the tumor microenvironment. The metabolic basis of how gas signaling molecules regulate angiogenesis is elusive. Herein, we report that hypoxic upregulation of NO synthesis in endothelial cells reprograms the transsulfuration pathway and increases H 2 S biogenesis. Furthermore, H 2 S oxidation by mitochondrial sulfide quinone oxidoreductase (SQOR) rather than downstream persulfides, synergizes with hypoxia to induce a reductive shift, limiting endothelial cell proliferation that is attenuated by dissipation of the mitochondrial NADH pool. Tumor xenografts in whole-body WB Cre SQOR fl/fl knockout mice exhibit lower mass and reduced angiogenesis compared to SQOR fl/fl controls. WB Cre SQOR fl/fl mice also exhibit reduced muscle angiogenesis following femoral artery ligation, compared to controls. Collectively, our data reveal the molecular intersections between H 2 S, O 2 and NO metabolism and identify SQOR inhibition as a metabolic vulnerability for endothelial cell proliferation and neovascularization. Highlights: Hypoxic induction of •NO in endothelial cells inhibits CBS and switches CTH reaction specificity Hypoxic interruption of the canonical transsulfuration pathway promotes H 2 S synthesis Synergizing with hypoxia, SQOR deficiency induces a reductive shift in the ETC and restricts proliferationSQOR KO mice exhibit lower neovascularization in tumor xenograft and hind limb ischemia models.

6.
Cell Metab ; 35(1): 134-149.e6, 2023 01 03.
Article in English | MEDLINE | ID: mdl-36528023

ABSTRACT

Effective therapies are lacking for patients with advanced colorectal cancer (CRC). The CRC tumor microenvironment has elevated metabolic waste products due to altered metabolism and proximity to the microbiota. The role of metabolite waste in tumor development, progression, and treatment resistance is unclear. We generated an autochthonous metastatic mouse model of CRC and used unbiased multi-omic analyses to reveal a robust accumulation of tumoral ammonia. The high ammonia levels induce T cell metabolic reprogramming, increase exhaustion, and decrease proliferation. CRC patients have increased serum ammonia, and the ammonia-related gene signature correlates with altered T cell response, adverse patient outcomes, and lack of response to immune checkpoint blockade. We demonstrate that enhancing ammonia clearance reactivates T cells, decreases tumor growth, and extends survival. Moreover, decreasing tumor-associated ammonia enhances anti-PD-L1 efficacy. These findings indicate that enhancing ammonia detoxification can reactivate T cells, highlighting a new approach to enhance the efficacy of immunotherapies.


Subject(s)
Ammonia , Colorectal Neoplasms , Animals , Mice , T-Cell Exhaustion , T-Lymphocytes , Colorectal Neoplasms/pathology , Immunotherapy , Tumor Microenvironment
7.
Cancer Cell ; 40(2): 185-200.e6, 2022 02 14.
Article in English | MEDLINE | ID: mdl-34951957

ABSTRACT

Microbial dysbiosis is a colorectal cancer (CRC) hallmark and contributes to inflammation, tumor growth, and therapy response. Gut microbes signal via metabolites, but how the metabolites impact CRC is largely unknown. We interrogated fecal metabolites associated with mouse models of colon tumorigenesis with varying mutational load. We find that microbial metabolites from healthy mice or humans are growth-repressive, and this response is attenuated in mice and patients with CRC. Microbial profiling reveals that Lactobacillus reuteri and its metabolite, reuterin, are downregulated in mouse and human CRC. Reuterin alters redox balance, and reduces proliferation and survival in colon cancer cells. Reuterin induces selective protein oxidation and inhibits ribosomal biogenesis and protein translation. Exogenous Lactobacillus reuteri restricts colon tumor growth, increases tumor reactive oxygen species, and decreases protein translation in vivo. Our findings indicate that a healthy microbiome and specifically, Lactobacillus reuteri, is protective against CRC through microbial metabolite exchange.


Subject(s)
Colorectal Neoplasms/metabolism , Colorectal Neoplasms/pathology , Gastrointestinal Microbiome , Glyceraldehyde/analogs & derivatives , Oxidation-Reduction , Propane/metabolism , Animals , Biomarkers , Cell Line, Tumor , Cell Proliferation/drug effects , Disease Models, Animal , Energy Metabolism , Glutathione/metabolism , Glyceraldehyde/metabolism , Glyceraldehyde/pharmacology , Host Microbial Interactions , Humans , Intestinal Mucosa/metabolism , Intestinal Mucosa/microbiology , Intestinal Mucosa/pathology , Metabolomics/methods , Metagenomics/methods , Mice , Models, Biological , Oxidation-Reduction/drug effects , Oxidative Stress , Propane/pharmacology , Signal Transduction , Xenograft Model Antitumor Assays
8.
Nat Metab ; 3(7): 969-982, 2021 07.
Article in English | MEDLINE | ID: mdl-34155415

ABSTRACT

Colorectal cancer (CRC) requires massive iron stores, but the complete mechanisms by which CRC modulates local iron handling are poorly understood. Here, we demonstrate that hepcidin is activated ectopically in CRC. Mice deficient in hepcidin specifically in the colon tumour epithelium, compared with wild-type littermates, exhibit significantly diminished tumour number, burden and size in a sporadic model of CRC, whereas accumulation of intracellular iron by deletion of the iron exporter ferroportin exacerbates these tumour parameters. Metabolomic analysis of three-dimensional patient-derived CRC tumour enteroids indicates a prioritization of iron in CRC for the production of nucleotides, which is recapitulated in our hepcidin/ferroportin mouse CRC models. Mechanistically, our data suggest that iron chelation decreases mitochondrial function, thereby altering nucleotide synthesis, whereas exogenous supplementation of nucleosides or aspartate partially rescues tumour growth in patient-derived enteroids and CRC cell lines in the presence of an iron chelator. Collectively, these data suggest that ectopic hepcidin in the tumour epithelium establishes an axis to sequester iron in order to maintain the nucleotide pool and sustain proliferation in colorectal tumours.


Subject(s)
Colorectal Neoplasms/metabolism , Hepcidins/metabolism , Iron/metabolism , Mitochondria/metabolism , Nucleotides/metabolism , Animals , Cell Line, Tumor , Epithelial Cells/metabolism , Humans , Mice
9.
JCI Insight ; 6(14)2021 06 17.
Article in English | MEDLINE | ID: mdl-34138755

ABSTRACT

Cancer cells reprogram cellular metabolism to maintain adequate nutrient pools to sustain proliferation. Moreover, autophagy is a regulated mechanism to break down dysfunctional cellular components and recycle cellular nutrients. However, the requirement for autophagy and the integration in cancer cell metabolism is not clear in colon cancer. Here, we show a cell-autonomous dependency of autophagy for cell growth in colorectal cancer. Loss of epithelial autophagy inhibits tumor growth in both sporadic and colitis-associated cancer models. Genetic and pharmacological inhibition of autophagy inhibits cell growth in colon cancer-derived cell lines and patient-derived enteroid models. Importantly, normal colon epithelium and patient-derived normal enteroid growth were not decreased following autophagy inhibition. To couple the role of autophagy to cellular metabolism, a cell culture screen in conjunction with metabolomic analysis was performed. We identified a critical role of autophagy to maintain mitochondrial metabolites for growth. Loss of mitochondrial recycling through inhibition of mitophagy hinders colon cancer cell growth. These findings have revealed a cell-autonomous role of autophagy that plays a critical role in regulating nutrient pools in vivo and in cell models, and it provides therapeutic targets for colon cancer.


Subject(s)
Colitis-Associated Neoplasms/immunology , Mitochondria/metabolism , Mitophagy/immunology , Nutrients/deficiency , Animals , Cell Line, Tumor , Cell Proliferation/drug effects , Colitis/chemically induced , Colitis/complications , Colitis/immunology , Colitis/pathology , Colitis-Associated Neoplasms/drug therapy , Colitis-Associated Neoplasms/genetics , Colitis-Associated Neoplasms/pathology , Colon/cytology , Colon/immunology , Colon/pathology , Dextran Sulfate/administration & dosage , Dextran Sulfate/toxicity , Disease Models, Animal , Female , Humans , Intestinal Mucosa/cytology , Intestinal Mucosa/immunology , Intestinal Mucosa/pathology , Male , Metabolomics , Mice , Mice, Transgenic , Mitochondria/immunology , Mitophagy/drug effects
10.
J Clin Invest ; 131(12)2021 06 15.
Article in English | MEDLINE | ID: mdl-33914705

ABSTRACT

Hypoxia is a hallmark of solid tumors that promotes cell growth, survival, and metastasis and confers resistance to chemo and radiotherapies. Hypoxic responses are largely mediated by the transcription factors hypoxia-inducible factor 1α (HIF-1α) and HIF-2α. Our work demonstrates that HIF-2α is essential for colorectal cancer (CRC) progression. However, targeting hypoxic cells is difficult, and tumors rapidly acquire resistance to inhibitors of HIF-2α. To overcome this limitation, we performed a small molecule screen to identify HIF-2α-dependent vulnerabilities. Several known ferroptosis activators and dimethyl fumarate (DMF), a cell-permeable mitochondrial metabolite derivative, led to selective synthetic lethality in HIF-2α-expressing tumor enteroids. Our work demonstrated that HIF-2α integrated 2 independent forms of cell death via regulation of cellular iron and oxidation. First, activation of HIF-2α upregulated lipid and iron regulatory genes in CRC cells and colon tumors in mice and led to a ferroptosis-susceptible cell state. Second, via an iron-dependent, lipid peroxidation-independent pathway, HIF-2α activation potentiated ROS via irreversible cysteine oxidation and enhanced cell death. Inhibition or knockdown of HIF-2α decreased ROS and resistance to oxidative cell death in vitro and in vivo. Our results demonstrated a mechanistic vulnerability in cancer cells that were dependent on HIF-2α that can be leveraged for CRC treatment.


Subject(s)
Basic Helix-Loop-Helix Transcription Factors/biosynthesis , Colorectal Neoplasms/metabolism , Gene Expression Regulation, Neoplastic , Iron/metabolism , Neoplasm Proteins/biosynthesis , Reactive Oxygen Species/metabolism , Animals , Basic Helix-Loop-Helix Transcription Factors/genetics , Cell Death/genetics , Cell Hypoxia/genetics , Colorectal Neoplasms/genetics , Colorectal Neoplasms/pathology , HCT116 Cells , HT29 Cells , Humans , Mice , Mice, Transgenic , Neoplasm Proteins/genetics , Oxidation-Reduction
11.
J Biol Chem ; 295(30): 10493-10505, 2020 07 24.
Article in English | MEDLINE | ID: mdl-32503843

ABSTRACT

The gastrointestinal tract is a highly proliferative and regenerative tissue. The intestine also harbors a large and diverse microbial population collectively called the gut microbiome (microbiota). The microbiome-intestine cross-talk includes a dynamic exchange of gaseous signaling mediators generated by bacterial and intestinal metabolisms. Moreover, the microbiome initiates and maintains the hypoxic environment of the intestine that is critical for nutrient absorption, intestinal barrier function, and innate and adaptive immune responses in the mucosal cells of the intestine. The response to hypoxia is mediated by hypoxia-inducible factors (HIFs). In hypoxic conditions, the HIF activation regulates the expression of a cohort of genes that promote adaptation to hypoxia. Physiologically, HIF-dependent genes contribute to the aforementioned maintenance of epithelial barrier function, nutrient absorption, and immune regulation. However, chronic HIF activation exacerbates disease conditions, leading to intestinal injury, inflammation, and colorectal cancer. In this review, we aim to outline the major roles of physiological and pathological hypoxic conditions in the maintenance of intestinal homeostasis and in the onset and progression of disease with a major focus on understanding the complex pathophysiology of the intestine.


Subject(s)
Adaptive Immunity , Colorectal Neoplasms , Gastrointestinal Microbiome/immunology , Hypoxia , Immunity, Innate , Oxygen/immunology , Animals , Basic Helix-Loop-Helix Transcription Factors/immunology , Colorectal Neoplasms/blood supply , Colorectal Neoplasms/immunology , Colorectal Neoplasms/microbiology , Colorectal Neoplasms/pathology , Humans , Hypoxia/immunology , Hypoxia/microbiology , Hypoxia/pathology , Inflammation/immunology , Inflammation/microbiology , Inflammation/pathology , Intestinal Mucosa/blood supply , Intestinal Mucosa/immunology , Intestinal Mucosa/microbiology , Intestinal Mucosa/pathology
12.
Blood Cells Mol Dis ; 77: 29-33, 2019 07.
Article in English | MEDLINE | ID: mdl-30953939

ABSTRACT

A young Indian female visited hospital as a suspected case of thrombotic thrombocytopenic purpura (TTP) with relapsed thrombotic complications with low platelet counts, infarct in middle cerebral artery and thrombi in microvessels. We first confirmed the deficiency of ADAMTS13 metalloprotease in this patient showing improper cleavage of vWF multimers by her plasma unlike her parents and brother. Although patient had very less ADAMTS13 antigen in plasma, but it did not appear to be the cause of deficiency of the enzyme, because her father had similarly low antigen level and he never had prothrombotic complications. While investigating the genetic change in ADAMTS13, we observed four homozygous-SNPs (g.420T>C, g.1342C>G, g.1716G>A and g.2280T>C) in exon 5, 12, 15 and 19 respectively in patient and her father unlike the heterozygous form of same SNPs in mother and brother. Further to investigate the cause of ADAMTS13 deficiency, we observed an elevated level of antibody against ADAMTS13 in patient unlike her father and other family members. Our study therefore provides the molecular approach of diagnosis of TTP in this patient and also highlights the use of such techniques in India. More importantly, study provides the clue of alternate treatment such as immunosuppressant therapy to this patient.


Subject(s)
ADAMTS13 Protein/immunology , Autoantibodies/immunology , Purpura, Thrombotic Thrombocytopenic/immunology , Purpura, Thrombotic Thrombocytopenic/metabolism , ADAMTS13 Protein/antagonists & inhibitors , ADAMTS13 Protein/blood , ADAMTS13 Protein/genetics , Autoantibodies/blood , Autoantigens/immunology , Autoimmunity , Enzyme Activation , Enzyme-Linked Immunosorbent Assay , Female , Humans , India , Male , Platelet Function Tests , Polymorphism, Genetic , Polymorphism, Single Nucleotide , Proteolysis , Purpura, Thrombotic Thrombocytopenic/diagnosis , Purpura, Thrombotic Thrombocytopenic/genetics , von Willebrand Factor/metabolism
13.
J Immunol Res ; 2019: 1409383, 2019.
Article in English | MEDLINE | ID: mdl-31032371

ABSTRACT

In a recent work, we have described the kinetics among the monocyte subsets in the peripheral blood of hemolytic patients including paroxysmal nocturnal hemoglobinuria (PNH) and sickle cell disease (SCD). After engulfing Hb-activated platelets, classical monocytes (CD14+CD16-) significantly transformed into highly inflammatory (CD14+CD16hi) subsets in vitro. An estimated 40% of total circulating monocytes in PNH and 70% in SCD patients existed as CD14+CD16hi subsets. In this study, we show that the nonclassical (CD14dimCD16+) monocyte subsets are nearly absent in patients with PNH or SCD, compared to 10-12% cells in healthy individuals. In mechanism, we have described the unique role of both free Hb and nitric oxide (NO) in reducing number of nonclassical subsets more than classical monocytes. After engulfing Hb-activated platelets, the monocytes including nonclassical subsets acquired rapid cell death within 12 h in vitro. Further, the treatment to monocytes either with the secretome of Hb-activated platelets containing NO and free Hb or purified free Hb along with GSNO (a physiological NO donor) enhanced rapid cell death. Besides, our data from both PNH and SCD patients exhibited a direct correlation between intracellular NO and cell death marker 7AAD in monocytes from the peripheral blood. Our data together suggest that due to the immune surveillance nature, the nonclassical or patrolling monocytes are encountered frequently by Hb-activated platelets, free Hb, and NO in the circulation of hemolytic patients and are predisposed to die rapidly.


Subject(s)
Hemoglobins/analysis , Hemoglobinuria, Paroxysmal/immunology , Monocytes/cytology , Nitric Oxide/blood , Adolescent , Adult , Apoptosis , Biomarkers/blood , Female , Humans , Male , Young Adult
14.
Eur J Immunol ; 48(8): 1285-1294, 2018 08.
Article in English | MEDLINE | ID: mdl-29677388

ABSTRACT

The distinct response shown by different phenotypes of macrophages and monocytes under various clinical conditions has put the heterogeneity of these cells into focus of investigation for several diseases. Recently, we have described that after engulfing hemoglobin (Hb)-activated platelets, classical monocytes differentiated into pro-inflammatory phenotypes, which were abundant in the circulation of paroxysmal nocturnal hemoglobinuria (PNH) and sickle cell disease patients. Our current study shows that upon engulfment of Hb-activated platelets, monocytes differentiate into M1-macrophages under M1-polarization stimulus (GM-CSF, IFN-γ + LPS). When grown under M2-polarization stimulus (M-CSF, IL-4 + IL13), the cells exhibited an M1-like phenotype, secreted elevated levels of pro-inflammatory cytokines including TNF-α and IL-1ß, and displayed loss of the secretion of cytokine such as IL-10 and also phagocytic ability unlike the conventional M2 macrophages. Interestingly, when differentiated under the above polarization stimulus, monocytes from PNH patients expressed high levels of CD80 and phospho-STAT1, like M1 macrophages. Hemolytic mice also exhibited a gradual increase in monocyte-platelet aggregates in circulation and accumulation of CD80high macrophages in thioglycollate-induced inflamed peritoneum. The spleen of the mice was also populated by CD80high macrophages with compromised phagocytic capacity. Our findings suggest that the hemolytic environment and specifically the Hb-activated platelets, which are abundant in circulation during intravascular hemolysis, closely regulate monocyte differentiation.


Subject(s)
Blood Platelets/immunology , Hemoglobins/metabolism , Hemoglobinuria, Paroxysmal/pathology , Hemolysis/immunology , Macrophages/cytology , Monocytes/cytology , Phagocytosis/immunology , Anemia, Sickle Cell/immunology , Anemia, Sickle Cell/pathology , Animals , B7-1 Antigen/metabolism , Disease Models, Animal , Hemoglobinuria, Paroxysmal/immunology , Humans , Interleukin-10/metabolism , Interleukin-1beta/metabolism , Mice , Mice, Inbred BALB C , STAT1 Transcription Factor/metabolism , Tumor Necrosis Factor-alpha/metabolism
15.
Clin Case Rep ; 6(4): 770-772, 2018 04.
Article in English | MEDLINE | ID: mdl-29636962

ABSTRACT

Blastic plasmacytoid dendritic cell neoplasm is an aggressive neoplasm with a median survival of only a few months despite treatment. An exhaustive immunohistochemical workup is required to differentiate it from myeloid sarcoma and extranodal NK/T cell lymphoma. Treatment is with induction using a regimen utilized for leukemia. Allogeneic hematopoietic stem cell transplantation is recommended for those who achieve remission following induction.

16.
Sci Rep ; 7: 41697, 2017 01 31.
Article in English | MEDLINE | ID: mdl-28139770

ABSTRACT

Thrombocytopenia is common in patients with dengue virus (DENV) infections. With a focus on understanding the possible mechanism of thrombocytopenia in DENV infections we described a direct correlation between activation and depletion of platelets in patients. Our data showed a sharp decrease in platelet counts at day 4 of fever in patients. The high DENV genome copies in platelets correlated directly with the elevated platelet activation along with increased binding of complement factor C3 and IgG on their surface at day 4. Recovery in platelet count was observed on day 10 through day 6 and 8 with simultaneous decrease in platelet activation markers. Further, our in vitro data supported the above observations describing a concentration-dependent increase in platelet activation by DENV serotype-2. The high copy number of DENV2 genome in the platelet pellet correlated directly with platelet activation, microparticle generation and clot formation. Furthermore the DENV2-activated platelets were phagocytosed in large numbers by the monocytes. The DENV2-mediated lysis and clearance of platelets were abrogated in presence of platelet activation inhibitor, prostacyclin. These observations collectively suggest that platelet activation status is an important determinant of thrombocytopenia in dengue infections. A careful strategy of inactivation of platelets may rescue them from rapid destruction during DENV infections.


Subject(s)
Dengue Virus , Dengue/complications , Platelet Activation , Thrombocytopenia/blood , Thrombocytopenia/etiology , Apoptosis/immunology , Blood Platelets/immunology , Blood Platelets/metabolism , Complement C3/immunology , Complement C3/metabolism , Dengue/virology , Dengue Virus/genetics , Genome, Viral , Humans , Immunoglobulin G/immunology , Immunoglobulin G/metabolism , Monocytes/immunology , Monocytes/metabolism , Phagocytosis/immunology , Platelet Activation/immunology , Platelet Count , Severity of Illness Index , Thrombocytopenia/diagnosis , Thrombosis/immunology , Viral Load
17.
Clin Immunol ; 175: 133-142, 2017 02.
Article in English | MEDLINE | ID: mdl-28039017

ABSTRACT

Monocytes and macrophage combat infections and maintain homeostatic balance by engulfing microbes and apoptotic cells, and releasing inflammatory cytokines. Studies have described that these cells develop anti-inflammatory properties upon recycling the free-hemoglobin (Hb) in hemolytic conditions. While investigating the phenotype of monocytes in two hemolytic disorders-paroxysmal nocturnal hemoglobinuria (PNH) and sickle cell disease (SCD), we observed a high number of pro-inflammatory (CD14+CD16hi) monocytes in these patients. We further investigated in vitro the phenotype of these monocytes and found an estimated 55% of CD14+ cells were transformed into the CD14+CD16hi subset after engulfing Hb-activated platelets. The CD14+CD16hi monocytes, which were positive for both intracellular Hb and CD42b (platelet marker), secreted significant amounts of TNF-α and IL-1ß, unlike monocytes treated with only free Hb, which secreted more IL-10. We have shown recently the presence of a high number of Hb-bound hyperactive platelets in patients with both diseases, and further investigated if the monocytes engulfed these activated platelets in vivo. As expected, we found 95% of CD14+CD16hi monocytes with both intracellular Hb and CD42b in both diseases, and they expressed high TNF-α. Furthermore our data showed that these monocytes whether from patients or developed in vitro after treatment with Hb-activated platelets, secreted significant amounts of tissue factor. Besides, these CD14+CD16hi monocytes displayed significantly decreased phagocytosis of E. coli. Our study therefore suggests that this alteration of monocyte phenotype may play a role in the increased propensity to pro-inflammatory/coagulant complications observed in these hemolytic disorders-PNH and SCD.


Subject(s)
Anemia, Sickle Cell/pathology , Blood Platelets/pathology , Hemoglobins/metabolism , Hemoglobinuria, Paroxysmal/pathology , Inflammation/pathology , Monocytes/pathology , Anemia, Sickle Cell/metabolism , Blood Platelets/metabolism , Hemoglobinuria, Paroxysmal/metabolism , Humans , Inflammation/metabolism , Interleukin-10/metabolism , Interleukin-1beta/metabolism , Lipopolysaccharide Receptors/metabolism , Macrophages/metabolism , Macrophages/pathology , Monocytes/metabolism , Phenotype , Platelet Glycoprotein GPIb-IX Complex/metabolism , Receptors, IgG/metabolism , Tumor Necrosis Factor-alpha/metabolism
18.
PLoS One ; 11(12): e0167899, 2016.
Article in English | MEDLINE | ID: mdl-27936141

ABSTRACT

Intravascular hemolysis increases the risk of thrombosis in hemolytic disorders. Our previous study showed that the binding of adult hemoglobin (HbA) to glycoprotein (GP) 1bα induced the activation of platelets. The elevated plasma Hb or platelet surface bound Hb positively correlated with platelet activation in patients with paroxysmal nocturnal hemoglobinuria (PNH). Furthermore, this study shows that the sickle Hb [HbS, occurs due to single nucleotide polymorphism at A>T of ß-globin gene of Hb and causes sickle cell disease (SCD)] also bound to GP1bα and activated platelets in a concentration-dependent manner. The HbS bound to glycocalicin (extramembranous part of GP1bα) with KD ~ 10.46 ± 3 µM. HbS induced phosphorylation of signaling adapter proteins, such as Lyn, PI3K, Akt and ERK in platelets, and also increased the surface expression of platelet activation markers such as P-selectin (10.7 fold) and PAC1 binding (10.4 fold) in platelet surface in a concentration-dependent manner. HbS also increased the platelet microparticle-generation (4.7 fold) and thrombus-formation (4.3 fold) in a concentration-dependent manner. An elevated level of extracellular Hb in plasma correlated directly with platelet activation markers such as P-selectin (r = 0.7947), PAC1 binding (r = 0.5914) on platelet surface and plasma levels of platelet-derived microparticles (r = 0.7834) in patients with SCD. Our study therefore suggests that the HbS-induced platelet activation may play a crucial role in intravascular clot formation observed in SCD patients characterized by high propensity to vascular occlusion and hypercoagulable states.


Subject(s)
Anemia, Sickle Cell/blood , Glycoproteins/metabolism , Hemoglobin, Sickle/metabolism , Immunoglobulins/metabolism , Platelet Activation , Enzyme-Linked Immunosorbent Assay , Humans , In Vitro Techniques , Microscopy, Confocal , Protein Binding , Surface Plasmon Resonance
19.
PLoS One ; 11(4): e0154276, 2016.
Article in English | MEDLINE | ID: mdl-27105433

ABSTRACT

BACKGROUND: Reports including our own describe that intravascular hemolysis increases the risk of thrombosis in hemolytic disorders. Our recent study shows that plasma Hb concentrations correlate directly with platelet activation in patients with paroxysmal nocturnal hemoglobinuria (PNH). The binding of Hb to glycoprotein1bα (GP1bα) increases platelet activation. A peptide AA1-50, designed from N-terminal amino acid sequence of GP1bα significantly inhibits the Hb binding to GP1bα as well as Hb-induced platelet activation. This study further examined if the Hb-mediated platelet activation plays any significant role in thrombus formation on subendothelium matrix under physiological flow shear stresses and the inhibition of Hb-platelet interaction can abrogate the above effects of Hb. METHODS AND RESULTS: Study performed thrombus formation assay in vitro by perfusing whole blood over immobilized VWF or collagen type I in presence of Hb under shear stresses simulating arterial or venous flow. The Hb concentrations ranging from 5 to 10 µM, commonly observed level in plasma of the hemolytic patients including PNH, dose-dependently increased thrombus formation on immobilized VWF under higher shear stress of 25 dyne/cm2, but not at 5 dyne/cm2. The above Hb concentrations also increased thrombus formation on immobilized collagen under both shear stresses of 5 and 25 dyne/cm2. The peptide AA1-50 abrogated invariably the above effects of Hb on thrombus formation. CONCLUSIONS AND SIGNIFICANCE: This study therefore indicates that the Hb-induced platelet activation plays a crucial role in thrombus formation on immobilized VWF or collagen under physiological flow shear stresses. Thus suggesting a probable role of this mechanism in facilitating thrombosis under hemolytic conditions.


Subject(s)
Collagen Type I/metabolism , Hemoglobins/metabolism , Platelet Glycoprotein GPIb-IX Complex/metabolism , Thrombosis/metabolism , von Willebrand Factor/metabolism , Amino Acid Sequence , Hemoglobinuria, Paroxysmal/blood , Hemoglobinuria, Paroxysmal/metabolism , Hemolysis , Humans , Immobilized Proteins/metabolism , Peptides/metabolism , Peptides/pharmacology , Platelet Activation/drug effects , Protein Binding/drug effects , Stress, Mechanical , Thrombosis/prevention & control
20.
Haematologica ; 100(12): 1526-33, 2015 Dec.
Article in English | MEDLINE | ID: mdl-26341739

ABSTRACT

Intravascular hemolysis increases the risk of hypercoagulation and thrombosis in hemolytic disorders. Our study shows a novel mechanism by which extracellular hemoglobin directly affects platelet activation. The binding of Hb to glycoprotein1bα activates platelets. Lower concentrations of Hb (0.37-3 µM) significantly increase the phosphorylation of signaling adapter proteins, such as Lyn, PI3K, AKT, and ERK, and promote platelet aggregation in vitro. Higher concentrations of Hb (3-6 µM) activate the pro-apoptotic proteins Bak, Bax, cytochrome c, caspase-9 and caspase-3, and increase platelet clot formation. Increased plasma Hb activates platelets and promotes their apoptosis, and plays a crucial role in the pathogenesis of aggregation and development of the procoagulant state in hemolytic disorders. Furthermore, we show that in patients with paroxysmal nocturnal hemoglobinuria, a chronic hemolytic disease characterized by recurrent events of intravascular thrombosis and thromboembolism, it is the elevated plasma Hb or platelet surface bound Hb that positively correlates with platelet activation.


Subject(s)
Apoptosis , Blood Platelets/metabolism , Hemoglobins/metabolism , Hemolysis , Platelet Activation , Platelet Glycoprotein GPIb-IX Complex/metabolism , Blood Platelets/pathology , Female , Hemoglobinuria, Paroxysmal/pathology , Humans , Male
SELECTION OF CITATIONS
SEARCH DETAIL