Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 92
Filter
1.
Blood Adv ; 5(21): 4380-4392, 2021 11 09.
Article in English | MEDLINE | ID: mdl-34500454

ABSTRACT

Homing and engraftment of hematopoietic stem/progenitor cells (HSPCs) into the bone marrow (BM) microenvironment are tightly regulated by the chemokine stromal cell-derived factor-1 (SDF-1) and its G-protein-coupled receptor C-X-C motif chemokine receptor 4 (CXCR4), which on engagement with G-protein subunits, trigger downstream migratory signals. Regulators of G-protein signaling (RGS) are GTPase-accelerating protein of the Gα subunit and R4 subfamily members have been implicated in SDF-1-directed trafficking of mature hematopoietic cells, yet their expression and influence on HSPCs remain mostly unknown. Here, we demonstrated that human CD34+ cells expressed multiple R4 RGS genes, of which RGS1, RGS2, RGS13, and RGS16 were significantly upregulated by SDF-1 in a CXCR4-dependent fashion. Forced overexpression of RGS1, RGS13, or RGS16 in CD34+ cells not only inhibited SDF-1-directed migration, calcium mobilization, and phosphorylation of AKT, ERK, and STAT3 in vitro, but also markedly reduced BM engraftment in transplanted NOD/SCID mice. Genome-wide microarray analysis of RGS-overexpressing CD34+ cells detected downregulation of multiple effectors with established roles in stem cell trafficking/maintenance. Convincingly, gain-of-function of selected effectors or ex vivo priming with their ligands significantly enhanced HSPC engraftment. We also constructed an evidence-based network illustrating the overlapping mechanisms of RGS1, RGS13, and RGS16 downstream of SDF-1/CXCR4 and Gαi. This model shows that these RGS members mediate compromised kinase signaling and negative regulation of stem cell functions, complement activation, proteolysis, and cell migration. Collectively, this study uncovers an essential inhibitory role of specific R4 RGS proteins in stem cell engraftment, which could potentially be exploited to develop improved clinical HSPC transplantation protocols.


Subject(s)
Hematopoietic Stem Cell Transplantation , RGS Proteins , Animals , Antigens, CD34 , Hematopoietic Stem Cells , Humans , Mice , Mice, Inbred NOD , Mice, SCID , RGS Proteins/genetics , Receptors, CXCR4/genetics
2.
FEBS Open Bio ; 11(7): 1907-1920, 2021 07.
Article in English | MEDLINE | ID: mdl-33932136

ABSTRACT

We previously demonstrated that microRNA(miR)-223 is overexpressed in intestinal tissue of infants with necrotizing enterocolitis (NEC). The objective of the current study was to identify the target gene of miR-223 and to investigate the role of the miR-223/nuclear factor I-A (NFIA) axis in cellular functions that underpin the pathophysiology of NEC. The target gene of miR-223 was identified by in silico target prediction bioinformatics, luciferase assay, and western blotting. We investigated downstream signals of miR-223 and cellular functions by overexpressing the miRNA in Caco-2 and FHs74 cells stimulated with lipopolysaccharide or lipoteichoic acid (LTA). NFIA was identified as a target gene of miR-223. Overexpression of miR-223 significantly induced MYOM1 and inhibited NFIA and RGN in Caco-2 cells, while costimulation with LTA decreased expression of GNA11, MYLK, and PRKCZ. Expression levels of GNA11, MYLK, IL-6, and IL-8 were increased, and levels of NFIA and RGN were decreased in FHs74 cells. These potential downstream genes were significantly correlated with levels of miR-223 or NFIA in primary NEC tissues. Overexpression of miR-223 significantly increased apoptosis of Caco-2 and FHs74 cells, while proliferation of FHs74 was inhibited. These results suggest that upon binding with NFIA, miR-223 regulates functional effectors in pathways of apoptosis, cell proliferation, G protein signaling, inflammation, and smooth muscle contraction. The miR-223/NFIA axis may play an important role in the pathophysiology of NEC by enhancing inflammation and tissue damage.


Subject(s)
Enterocolitis, Necrotizing , MicroRNAs , Caco-2 Cells , Enterocolitis, Necrotizing/genetics , Enterocolitis, Necrotizing/metabolism , Humans , Infant, Newborn , Inflammation/genetics , MicroRNAs/genetics , MicroRNAs/metabolism , NFI Transcription Factors/genetics , NFI Transcription Factors/metabolism
3.
Neonatology ; 117(5): 555-561, 2020.
Article in English | MEDLINE | ID: mdl-33238267

ABSTRACT

OBJECTIVE: The objective of this study is to evaluate the usefulness of fecal microRNA (miR)-223 and miR-451a, as novel noninvasive biomarkers for early diagnosis of necrotizing enterocolitis (NEC) in preterm infants. METHODS: Among the top-listed target miRNAs in our previous differential microarray analysis, miR-223 and miR-451a were quantified in a pilot validation case-controlled study (NEC vs. non-NEC/nonsepsis infants; n = 6 in each group). A definitive prospective cohort study (n = 218) further assessed their clinical usefulness as noninvasive and specific diagnostic biomarkers. Fecal calprotectin was quantified in parallel for comparison. RESULTS: Of 43 proven NEC cases in the cohort study, 24 (55.8%) had fecal samples recovered within the first 3 days of clinical presentation. Fecal miRNA-223 (10.5 fold), miR-451a (4.5 fold), and calprotectin (2.1 fold) concentrations were significant higher in NEC compared with the non-NEC group (p < 0.009). Accepting a minimum sensitivity of 0.75, the positive predictive values (PPVs) ranged between 0.19 and 0.20. Combining fecal biomarkers and CRP (Day 1) could marginally increase the PPVs (0.31-0.34) but adversely lowered the sensitivity (0.54-0.63). CONCLUSIONS: Although fecal miRNA biomarkers and calprotectin concentrations were significantly higher in the NEC group, the considerable overlapping of concentrations between groups and low recovery of stool specimens within 72 h of clinical presentation rendered fecal noninvasive tests of limited clinical value in guiding diagnosis of NEC during the acute phase. A further study is underway to evaluate their roles in surveillance for predicting high-risk premature infants developing NEC.


Subject(s)
Enterocolitis, Necrotizing , MicroRNAs , Biomarkers , Cohort Studies , Humans , Infant , Infant, Newborn , Infant, Premature , Prospective Studies
5.
Leukemia ; 34(3): 709-720, 2020 03.
Article in English | MEDLINE | ID: mdl-31624373

ABSTRACT

CD9 has been implicated in cancer progression but its prognostic relevance and therapeutic potential in B-cell precursor acute lymphoblastic leukemia (BCP-ALL) are largely unknown. In a cohort of pediatric BCP-ALL patients, we found that CD9+ cases had a significantly lower 5-year relapse-free survival rate than CD9- cases. Multivariate analysis demonstrated that CD9 positivity independently predicted inferior survival outcomes, and could be applied with established prognostic features, including prednisone response and cytogenetic status, to refine patient stratification. Administration of CD9 antibody substantially suppressed disease progression in NOD/SCID mice xenografted with CD9+ cell lines and primary leukemic blasts from patients with high-risk and refractory BCP-ALL, without compromising hematopoietic stem cell engraftment. Combination of anti-CD9 with conventional chemotherapy further reduced leukemic burden and prolonged animal survival. Mechanistically, CD9 blockade inhibited leukemic cell proliferation, induced G0/G1 cell cycle arrest, activated p38, and enhanced chemotherapeutic agent-induced apoptosis. Further, CD9 physically interacted with integrin very late antigen-4, regulated affinity to vascular cell adhesion molecule-1, and was involved in leukemia-stroma interaction. Collectively, our study established CD9 as a new prognostic marker, validated the preclinical efficacy of CD9 antibody, and laid the foundation for clinical development of CD9-targeted therapy for high-risk and refractory pediatric BCP-ALL.


Subject(s)
Precursor B-Cell Lymphoblastic Leukemia-Lymphoma/drug therapy , Tetraspanin 29/antagonists & inhibitors , Animals , Cell Cycle , Cell Line, Tumor , Cell Lineage , Child , Disease Progression , Disease-Free Survival , Hematopoietic Stem Cells/cytology , Humans , Immunophenotyping , Mice , Mice, Inbred NOD , Mice, SCID , Multivariate Analysis , Neoplasm Transplantation , Precursor B-Cell Lymphoblastic Leukemia-Lymphoma/pathology , Prognosis , Treatment Outcome , p38 Mitogen-Activated Protein Kinases/metabolism
6.
FASEB J ; 33(4): 5143-5152, 2019 04.
Article in English | MEDLINE | ID: mdl-30624964

ABSTRACT

The level of microRNA (miR)-431 was found to be markedly up-regulated in intestinal tissue of necrotizing enterocolitis (NEC). The objective of this study was to identify the target gene of miR-431 and to investigate the role of the miR-431-FOXA1 axis in the pathophysiology of NEC. The target gene of miR-431 was identified by in silico target prediction bioinformatics, luciferase assay, and Western blotting. Effects of miR-431 on downstream expression signals, cell proliferation, and apoptosis were investigated by overexpression in Caco-2 cells upon stimulation by LPS or lipoteichoic acid (LTA). FOXA1 was identified as the target gene of miR-431. Overexpression of miR-431 in Caco-2 cells significantly inhibited FOXA1, ESRRG, and HNF4A and activated IL-6, LGR5, NFKB2, PLA2G2A, PRKCZ, and TNF. IL-8 and - 10 were enhanced when costimulated with LPS or LTA. These potential downstream genes were also significantly dysregulated in primary NEC tissues compared with surgical-control tissues. Overexpression of miR-431 significantly decreased proliferation and increased apoptosis of Caco-2 cells. A proposed network of miR-431-FOXA1 interaction with LPS and LTA receptors demonstrates dysregulation of transcription factors, inflammatory mediators, epithelium tight junction regulators, and cell proliferation and apoptosis signals. The miR-431-FOXA1 axis could in part be responsible for the intensification of the inflammatory response in NEC tissues and contribute to the proinflammatory pathophysiology.-Wu, Y. Z., Chan, K. Y. Y., Leung, K. T., Lam, H. S., Tam, Y. H., Lee, K. H., Li, K., Ng, P. C. Dysregulation of miR-431 and target gene FOXA1 in intestinal tissues of infants with necrotizing enterocolitis.


Subject(s)
Enterocolitis, Necrotizing/metabolism , Hepatocyte Nuclear Factor 3-alpha/metabolism , MicroRNAs/metabolism , Apoptosis/drug effects , Apoptosis/genetics , Blotting, Western , Caco-2 Cells , Cell Proliferation/drug effects , Cell Proliferation/genetics , Enterocolitis, Necrotizing/genetics , Flow Cytometry , HEK293 Cells , Hepatocyte Nuclear Factor 3-alpha/genetics , Humans , Intestinal Mucosa/metabolism , Lipopolysaccharides/pharmacology , MicroRNAs/genetics , Plasmids/genetics , Real-Time Polymerase Chain Reaction , Teichoic Acids/pharmacology
7.
J Pediatr ; 205: 83-90.e10, 2019 02.
Article in English | MEDLINE | ID: mdl-30529132

ABSTRACT

OBJECTIVE: To discover specific circulating microRNA (miRNA) biomarkers for the early differentiation of necrotizing enterocolitis (NEC) from neonatal sepsis and inflammatory conditions. STUDY DESIGN: The study comprised 3 distinct phases: differential microarray analysis to compare plasma miRNA expression profiles of NEC vs sepsis and non-NEC/nonsepsis cases, a case-control study to quantify dysregulated miRNAs as potential specific biomarkers of NEC, and a prospective cohort study to assess the diagnostic usefulness of the best miRNA biomarker(s). RESULTS: A distinct miRNA expression profile was observed in the NEC compared with the sepsis and non-NEC/nonsepsis groups. miR-1290, miR-1246, and miR-375 were discovered to be specific biomarkers of NEC in the case-control study. In the cohort study (n = 301), plasma miR-1290 (day 0; >220 copies/µL) provided the greatest diagnostic usefulness for identifying both mild medical and severe surgical NEC cases. Of 20 infants with miR-1290 >650 copies/µL, 15 were diagnosed with NEC. Incorporating C-reactive protein (day 1; >15.8 mg/L) for cases with intermediate levels (220-650 copies/µL) in a 2-stage algorithm further optimized the diagnostic profile with a sensitivity of 0.83, a specificity of 0.96, a positive predictive value of 0.75, and a negative predictive value of 0.98. Importantly, 7 of 36 infants with NEC (19.4%) could be diagnosed 7.8-32.2 hours earlier (median, 13.3 hours) using miR-1290. CONCLUSIONS: Plasma miR-1290 is a novel and specific biomarker that can effectively differentiate NEC cases from neonatal sepsis. miR-1290 facilitates neonatologists to confidently and timely reach a decision for early transfer of sick infants with NEC from community-based hospitals to tertiary surgical centers.


Subject(s)
Enterocolitis, Necrotizing/blood , MicroRNAs/blood , Biomarkers/blood , C-Reactive Protein/analysis , Case-Control Studies , Diagnosis, Differential , Early Diagnosis , Enterocolitis, Necrotizing/diagnosis , Enterocolitis, Necrotizing/genetics , Female , Gestational Age , Humans , Infant , Infant, Newborn , Male , Microarray Analysis , Neonatal Sepsis/diagnosis , Predictive Value of Tests , Prospective Studies
8.
Neonatology ; 115(3): 189-196, 2019.
Article in English | MEDLINE | ID: mdl-30580336

ABSTRACT

Recent advances in molecular and mass screening technologies have paved the way for discovery of novel diagnostic tests and/or biomarkers for accurate identification of specific diseases and organ injuries. However, new diagnostic tests/biomarkers should be subjected to thorough evaluation before introduction into routine clinical practice. This review focuses on the up-to-date methodology in designing and evaluating diagnostic tests/biomarkers, and assessing their clinical utilities for improving health care efficiency, cost-effectiveness and outcomes. In addition to improved diagnostic utilities, future diagnostic tests should be developed in collaboration with our industrial partners and be applicable at the bedside for disease surveillance.


Subject(s)
Biomarkers/analysis , Diagnostic Tests, Routine/standards , Diagnostic Tests, Routine/trends , Cooperative Behavior , Cost-Benefit Analysis , Humans , Research Design
9.
Semin Fetal Neonatal Med ; 23(6): 380-386, 2018 12.
Article in English | MEDLINE | ID: mdl-30082194

ABSTRACT

Necrotizing enterocolitis (NEC) remains a devastating surgical emergency with high morbidity and mortality in preterm infants. Slow but steady progress has been made in past years searching for novel biomarkers of NEC, for both surveillance and diagnostic purposes. This review primarily focuses on recent discoveries: clinical applications of different categories of biomarkers for surveillance, early diagnosis, and predicting severity and prognosis; and understanding of pathophysiological mechanisms as a basis to rationalize the search for 'gut-associated specific biomarkers' of NEC. An important next step is to collaborate with our industrial partners to develop point-of-care tests, and to discover novel and gut-associated specific biomarkers that can be used for surveillance and early diagnosis of NEC in routine clinical settings.


Subject(s)
Biomarkers , Enterocolitis, Necrotizing/diagnosis , Early Diagnosis , Humans , Infant, Newborn , Infant, Premature , Prognosis
10.
Eur J Immunol ; 48(6): 1059-1073, 2018 06.
Article in English | MEDLINE | ID: mdl-29460338

ABSTRACT

Endometriosis affects women of reproductive age via unclear immunological mechanism(s). Myeloid-derived suppressor cells (MDSCs) are a heterogeneous group of myeloid cells with potent immunosuppressive and angiogenic properties. Here, we found MDSCs significantly increased in the peripheral blood of patients with endometriosis and in the peritoneal cavity of a mouse model of surgically induced endometriosis. Majority of MDSCs were granulocytic, produced ROS, and arginase, and suppressed T-cell proliferation. Depletion of MDSCs by antiGr-1 antibody dramatically suppressed development of endometrial lesions in mice. The chemokines CXCL1, 2, and 5 were expressed at sites of lesion while MDSCs expressed CXCR-2. These CXC-chemokines promoted MDSC migration toward endometriotic implants both in vitro and in vivo. Also, CXCR2-deficient mice show significantly decreased MDSC induction, endometrial lesions, and angiogenesis. Importantly, adoptive transfer of MDSCs into CXCR2-KO mice restored endometriotic growth and angiogenesis. Together, this study demonstrates that MDSCs play a role in the pathogenesis of endometriosis and identifies a novel CXC-chemokine and receptor for the recruitment of MDSCs, thereby providing a potential target for endometriosis treatment.


Subject(s)
Angiogenesis Inducing Agents/immunology , Endometriosis/immunology , Endometrium/immunology , Granulocytes/immunology , Myeloid-Derived Suppressor Cells/immunology , Animals , Arginase/metabolism , Cells, Cultured , Chemokines/metabolism , Cytokines/metabolism , Disease Models, Animal , Endometrium/blood supply , Female , Humans , Mice , Mice, Inbred C57BL , Mice, Knockout , Reactive Oxygen Species/metabolism , Receptors, Interleukin-8B/genetics , Receptors, Interleukin-8B/metabolism
11.
Neonatology ; 111(2): 145-152, 2017.
Article in English | MEDLINE | ID: mdl-27756063

ABSTRACT

BACKGROUND: Neonatal sepsis remains an important cause of neonatal morbidity and mortality. Tools to rapidly predict antibiotic resistance in neonatal sepsis would be extremely valuable. OBJECTIVES: To develop quantitative polymerase chain reaction (qPCR) primer/probe sets that can rapidly detect antibiotic resistance genes common to a neonatal unit, and to investigate the feasibility of direct detection of antibiotic resistance genes in whole blood of infants with Gram-negative septicaemia without first isolating the organism. METHODS: Primer/probe sets were designed to detect genes that produce aminoglycoside-modifying enzymes or extended-spectrum ß-lactamase. In phase 1, Gram-negative organisms isolated from neonatal clinical specimens within a 12-month period were analysed by qPCR to detect preselected genes. In phase 2, blood specimens of infants with Gram-negative septicaemia were subjected to qPCR analysis to detect antibiotic resistance genes for comparison against conventional antibiotic resistance profile results. RESULTS: Two primer/probe sets showed promising diagnostic utilities for the prediction of antibiotic resistance; the diagnostic utilities (sensitivity, specificity, positive predictive value and negative predictive value) were 90.9, 96.4, 92.6 and 95.5%, respectively, for AAC3-2 [aac(3')-IIa/aacC3/aacC2, aac(3')-IIc/aacC2] to detect gentamicin resistance, and 59.3, 99.3, 94.1 and 92.6%, respectively, for BLA-C1 (blaCTX-M-9, blaCTX-M-14, blaCTX-M-24, blaCTX-M-27) to detect cephalosporin resistance. Twenty-six infants were tested in phase 2, and both gentamicin and cephalosporin resistance patterns were predicted with 100% sensitivity and 100% specificity by AAC3-2 and BLA-C1, respectively. CONCLUSIONS: qPCR with appropriately designed primer/probe sets can predict antibiotic resistance directly from neonatal blood, and it can substantially reduce the turnaround time for antibiotic resistance results.


Subject(s)
Drug Resistance, Bacterial/genetics , Gram-Negative Bacteria/genetics , Gram-Negative Bacteria/isolation & purification , Gram-Negative Bacterial Infections/diagnosis , Neonatal Sepsis/complications , Anti-Bacterial Agents/therapeutic use , Gram-Negative Bacteria/drug effects , Gram-Negative Bacterial Infections/drug therapy , Hong Kong , Humans , Infant, Newborn , Microbial Sensitivity Tests , Neonatal Sepsis/microbiology , Predictive Value of Tests
12.
Stem Cells Dev ; 25(23): 1780-1787, 2016 12 01.
Article in English | MEDLINE | ID: mdl-27596606

ABSTRACT

Preterm infants are at high risk of developing severe sepsis. Circulating hematopoietic stem and progenitor cells (HSPCs; CD45+CD34+) have been suggested to play a vital role in the host immunological defense against invading pathogens. The objectives were to investigate the regulation of circulating HSPCs in preterm infants during infection episodes, and to assess the relationship of CD45+CD34+ cells with immunological mediators and differential leukocyte populations. First, we conducted a cross-sectional case-control study comparing these parameters among infected infants (n = 23), gestational and postnatal age-matched noninfected infants (n = 46), and "healthy" control (CTL) infants (n = 12). Second, we investigated the longitudinal change of CD45+CD34+ cell concentrations in infected infants before, during, and after an infection episode, and compared them with the other two groups. Our cross-sectional results showed that CD45+CD34+ cell count and percentage were significantly reduced in infected infants during systemic infection, compared with the noninfected or CTL infants. There were significant positive correlation between levels of CD45+CD34+ cells and lymphocytes or monocytes, and significant negative correlation between CD45+CD34+ cells and neutrophils or interleukin (IL)-6 in infected infants. Longitudinal analysis showed that changes of CD45+CD34+ cells at the onset of sepsis relative to levels 1 week prior and 1 week postsepsis in infected infants were significantly different from those changes in the corresponding time points for the other two groups. Our findings suggested that circulating HSPCs were dynamically regulated during septicemia and could play an important role in the defense mechanism, plausibly contributing to replenishment of leukocytes during sepsis in preterm infants.


Subject(s)
Cell Movement , Hematopoietic Stem Cells/cytology , Infant, Premature/physiology , Sepsis/pathology , Antigens, CD/metabolism , Case-Control Studies , Female , Granulocyte Colony-Stimulating Factor/metabolism , Humans , Infant, Newborn , Interleukin-6/metabolism , Male , Sepsis/immunology , T-Lymphocytes, Cytotoxic/immunology
13.
Arch Dis Child Fetal Neonatal Ed ; 101(6): F571-F576, 2016 Nov.
Article in English | MEDLINE | ID: mdl-27601464

ABSTRACT

Preterm, very low birthweight (VLBW) infants are prone to life-threatening hypotension secondary to hypothalamic-pituitary-adrenal axis immaturity, resulting in adrenocortical insufficiency. Clinical presentations of inotrope-resistant refractory hypotension are usually evident, but interpretation of serum cortisol may pose much difficulty to front-line neonatologists. This review examines the salient pathophysiology of adrenocortical insufficiency in the immediate postnatal period, characterises its endocrinological abnormalities, and describes the typical and variant clinical presentations. Based on existing evidence, a practical scheme is proposed for logical interpretation of circulating cortisol levels and management of inotrope-resistant refractory hypotension in VLBW infants.

14.
J Pediatr ; 177: 78-83.e3, 2016 10.
Article in English | MEDLINE | ID: mdl-27480200

ABSTRACT

OBJECTIVES: To assess preferences of health care workers (HCWs) and parents of term and preterm infants to adverse health outcomes, and how perceived surgical mortality influences decision-making. STUDY DESIGN: A total of 536 participants (157 HCWs, 201 parents of term infants, and 178 parents of preterm infants) were recruited to take part in a structured interview. Preferences related to treatment of a critically ill preterm infant with necrotizing enterocolitis were measured by health state rank permutation analysis and pivotal risk analysis. Between-group and subgroup comparisons were performed. RESULTS: HCWs rank adverse health states less favorably than parents of preterm infants, consistently ranking 2 of the most adverse health states worse than death. Pivotal risk values of HCWs for all health states were consistently the lowest of the 3 groups. High operative mortality was associated uniformly with reduction in pivotal risks for all groups both in favorable and adverse health states. Subgroup analyses revealed significant discrepancies in preferences between fathers and mothers as well as doctors and nurses. Regular religious practice was significantly associated with increased pivotal risks in parental subgroups. CONCLUSIONS: As discrepancies in health state preferences existed between subgroups (ie, doctors vs nurses, mothers vs fathers) and perceived operative mortality consistently biased parental and HCW health state preferences, we recommend that HCWs should first identify differences regarding patient management before interviewing the parents together. HCWs should be aware of inadvertently biasing parents when discussing the risks and outcomes of surgery in conjunction with the overall long-term prognosis of the underlying condition.


Subject(s)
Attitude of Health Personnel , Decision Making , Parents/psychology , Surgical Procedures, Operative/psychology , Female , Hong Kong , Humans , Infant, Newborn , Infant, Premature , Male , Risk Factors , Term Birth
15.
Neonatology ; 110(4): 267-269, 2016.
Article in English | MEDLINE | ID: mdl-27304999

ABSTRACT

Neonatal haemophagocytic lymphohistiocytosis (HLH) is a rare but potentially lethal condition. We recently encountered a preterm infant who developed severe HLH associated with maternal adult-onset Still's disease, which to our knowledge has not been previously reported. The infant presented with fever, generalised lymphadenopathy, transient erythematous skin rash, hepatosplenomegaly, ascites, pancytopenia, marked hyperferritinaemia, and hypofibrinogenaemia, which were features similar to maternal presentation during late pregnancy. Whole gene exome sequencing screening for familial HLH (PRF1, STX11, STXBP2, and MUNC13D genes) was negative. We postulated that factors such as auto-antibodies, antigens, or inflammatory mediators transmitted vertically from the mother could have triggered the intense inflammation in the infant. The infant responded promptly to dexamethasone, etoposide, and cyclosporin A, without the need for bone marrow transplantation. Neonatologists should be alerted to the rare diagnosis of HLH in the presence of active maternal diseases, including infection or autoimmune conditions, especially in association with fever, cytopenia, and hepatosplenomegaly.


Subject(s)
Infant, Premature, Diseases/diagnosis , Lymphohistiocytosis, Hemophagocytic/diagnosis , Pregnancy Complications/diagnosis , Still's Disease, Adult-Onset/diagnosis , Cyclosporine/administration & dosage , Etoposide/administration & dosage , Female , Fever , Genetic Testing , Humans , Infant , Infant, Newborn , Infant, Premature , Infant, Premature, Diseases/drug therapy , Lymphohistiocytosis, Hemophagocytic/drug therapy , Lymphohistiocytosis, Hemophagocytic/genetics , Pregnancy , Prenatal Diagnosis
16.
J Nutr Biochem ; 29: 64-72, 2016 Mar.
Article in English | MEDLINE | ID: mdl-26895666

ABSTRACT

The small intestine is the exclusive site of arginine synthesis in neonates. Low levels of circulating arginine have been associated with the occurrence of necrotizing enterocolitis (NEC) but the mechanism of arginine dysregulation has not been fully elucidated. We aimed to investigate (i) expressional changes of arginine synthesizing and catabolic enzymes in human intestinal tissues of NEC, spontaneous intestinal perforation (SIP) and noninflammatory surgical conditions (Surg-CTL) and to investigate the (ii) mechanisms of arginine dysregulation and enterocyte proliferation upon stimulation by bacterial components, arginine depletion, ARG1 overexpression and nitric oxide (NO) supplementation. Our results showed that expressions of arginine synthesizing enzymes ALDH18A1, ASL, ASS1, CPS1, GLS, OAT and PRODH were significantly decreased in NEC compared with Surg-CTL or SIP tissues. Catabolic enzyme ARG1 was increased (>100-fold) in NEC tissues and histologically demonstrated to be expressed by infiltrating neutrophils. No change in arginine metabolic enzymes was observed between SIP and Surg-CTL tissues. In CaCO2 cells, arginine metabolic enzymes were differentially dysregulated by lipopolysaccharide or lipoteichoic acid. Depletion of arginine reduced cell proliferation and this phenomenon could be partially rescued by NO. Overexpression of ARG1 also reduced enterocyte proliferation. We provided the first expressional profile of arginine metabolic enzymes at the tissue level of NEC. Our findings suggested that arginine homeostasis was severely disturbed and could be triggered by inflammatory responses of enterocytes and infiltrating neutrophils as well as bacterial components. Such reactions could reduce arginine and NO, resulting in mucosal damage. The benefit of arginine supplementation for NEC prophylaxis merits further clinical evaluation.


Subject(s)
Arginine/metabolism , Enterocolitis, Necrotizing/enzymology , Intestines/enzymology , Arginase/genetics , Arginine/biosynthesis , Caco-2 Cells , Female , Humans , Infant , Infant, Newborn , Intestines/microbiology , Male
17.
PLoS One ; 10(8): e0135737, 2015.
Article in English | MEDLINE | ID: mdl-26274503

ABSTRACT

BACKGROUND: Necrotizing enterocolitis (NEC) and spontaneous intestinal perforation (SIP) are acute intestinal conditions which could result in mortality and severe morbidity in preterm infants. Our objective was to identify dysregulated micro-RNAs (miRNAs) in small bowel tissues of NEC and SIP, and their possible roles in disease pathophysiology. METHODS: We performed differential miRNA arrays on tissues of NEC (n = 4), SIP (n = 4) and surgical-control (Surg-CTL; n = 4), and validated target miRNAs by qPCR (n = 10 each group). The association of target miRNAs with 52 dysregulated mRNAs was investigated by bioinformatics on functional and base-pair sequence algorithms, and correlation in same tissue samples. RESULTS: We presented the first miRNA profiles of NEC, SIP and Surg-CTL intestinal tissues in preterm infants. Of 28 validated miRNAs, 21 were significantly different between NEC or SIP and Surg-CTL. Limited overlapping in the aberrant expression of miRNAs between NEC and SIP indicated their distinct molecular mechanisms. A proposed network of dysregulated miRNA/mRNA pairs in NEC suggested interaction at bacterial receptor TLR4 (miR-31, miR-451, miR-203, miR-4793-3p), mediated via key transcription factors NFKB2 (miR-203), AP-1/FOSL1 (miR-194-3p), FOXA1 (miR-21-3p, miR-431 and miR-1290) and HIF1A (miR-31), and extended downstream to pathways of angiogenesis, arginine metabolism, cell adhesion and chemotaxis, extracellular matrix remodeling, hypoxia/oxidative stress, inflammation and muscle contraction. In contrast, upregulation of miR-451 and miR-223 in SIP suggested modulation of G-protein-mediated muscle contraction. CONCLUSIONS: The robust response of miRNA dysregulation in NEC and SIP, and concerted involvement of specific miRNAs in the molecular networks indicated their crucial roles in mucosa integrity and disease pathophysiology.


Subject(s)
Enterocolitis, Necrotizing/metabolism , Gene Expression Regulation , Intestinal Mucosa/metabolism , Intestinal Perforation/metabolism , Intestine, Small/metabolism , MicroRNAs/biosynthesis , Enterocolitis, Necrotizing/pathology , Female , Gene Expression Profiling , Humans , Infant, Newborn , Infant, Premature , Intestinal Mucosa/pathology , Intestinal Perforation/pathology , Intestine, Small/pathology , Male
18.
World J Clin Pediatr ; 4(3): 45-9, 2015 Aug 08.
Article in English | MEDLINE | ID: mdl-26261766

ABSTRACT

AIM: To review the clinical response to levetiracetam (LEV) in neonatal seizure management in intensive care unit. METHODS: Medical records of neonates who received LEV from January 2009 to August 2014 were reviewed. Their demographic data, clinical characteristics, etiology, seizures, electroencephalograms, response to treatment and outcome were noted. Literature review of use of LEV in neonates were also performed via PubMed and EMBASE with keywords - "neonates", "seizures", "epilepsy" and "LEV" up to Sep 2014 and retrieved the publications. The response rate to LEV was compared. RESULTS: Twelve neonates were identified during the study period. All patients received phenobarbitone loading prior to consideration of LEV. Seven (58%) and nine (75%) achieved seizure freedom 24 h and 72 h after LEV was added, both clinically and electrographically. No serious adverse effects were associated with LEV use. From the literature, there are total 144 neonates reported to have used LEV. The overall results suggested that LEV could control up to 90% of neonatal seizures. CONCLUSION: LEV was found to be relatively safe and efficacious in treating neonatal seizures, but might not work well in the most severe hypoxic ischemic encephalopathy.

19.
Arch Dis Child Fetal Neonatal Ed ; 100(5): F448-52, 2015 Sep.
Article in English | MEDLINE | ID: mdl-25555389

ABSTRACT

Biomarkers have been used to differentiate systemic neonatal infection and necrotising enterocolitis (NEC) from other non-infective neonatal conditions that share similar clinical features. With increasing understanding in biochemical characteristics of different categories of biomarkers, a specific mediator or a panel of mediators have been used in different aspects of clinical management in neonatal sepsis/NEC. This review focuses on how these biomarkers can be used in real-life clinical settings for daily surveillance, bedside point-of-care testing, early diagnosis and predicting the severity and prognosis of neonatal sepsis/NEC. In addition, with recent development of 'multi-omic' approaches and rapid advancement in knowledge of bioinformatics, more novel biomarkers and unique signatures of mediators would be discovered for diagnosis of specific diseases and organ injuries.


Subject(s)
Biomarkers , Enterocolitis, Necrotizing/diagnosis , Infant, Premature, Diseases/diagnosis , Sepsis/diagnosis , Early Diagnosis , Humans , Infant, Newborn , Infant, Premature , Point-of-Care Systems , Prognosis , Severity of Illness Index
20.
PLoS One ; 9(3): e92706, 2014.
Article in English | MEDLINE | ID: mdl-24675897

ABSTRACT

Makorin-2 (MKRN2) is a highly conserved protein and yet its functions are largely unknown. We investigated the expression levels of MKRN2 and RAF1 in normal and malignant hematopoietic cells, and leukemia cell lines. We also attempted to delineate the role of MKRN2 in umbilical cord blood CD34+ stem/progenitor cells and K562 cell line by over-expression and inhibition of MKRN2 through lentivirus transduction and shRNA nucleofection, respectively. Our results provided the first evidence on the ubiquitous expression of MKRN2 in normal hematopoietic cells, embryonic stem cell lines, primary leukemia and leukemic cell lines of myeloid, lymphoid, erythroid and megakaryocytic lineages. The expression levels of MKRN2 were generally higher in primary leukemia samples compared with those in age-matched normal BM cells. In all leukemia subtypes, there was no significant correlation between expression levels of MKRN2 and RAF1. sh-MKRN2-silenced CD34+ cells had a significantly lower proliferation capacity and decreased levels of the early stem/progenitor subpopulation (CFU-GEMM) compared with control cultures. Over-expression of MKRN2 in K562 cells increased cell proliferation. Our results indicated possible roles of MKRN2 in normal and malignant hematopoiesis.


Subject(s)
Gene Expression , Hematopoiesis/genetics , Leukemia/genetics , Leukocytes, Mononuclear/metabolism , Ribonucleoproteins/genetics , Cell Line, Tumor , Cells, Cultured , Embryonic Stem Cells/metabolism , Gene Silencing , Hematopoietic Stem Cells/metabolism , Humans , K562 Cells , Proto-Oncogene Proteins c-raf/genetics , Proto-Oncogene Proteins c-raf/metabolism , RNA, Messenger/genetics , RNA, Small Interfering/genetics , Ribonucleoproteins/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL