Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 27
Filter
2.
J Chemother ; 21(3): 322-9, 2009 Jun.
Article in English | MEDLINE | ID: mdl-19567354

ABSTRACT

The aims of the study were to analyze the clinical and epidemiological characteristics and treatments for patients who developed zygomycosis enrolled in Italy during the European Confederation of Medical Mycology of medical mycology survey. This prospective multicenter study was performed between 2004 and 2007 at 49 italian Departments. 60 cases of zygomycosis were enrolled: the median age was 59.5 years (range 1-87), with a prevalence of males (70%). The majority of cases were immunocompromised patients (42 cases, 70%), mainly hematological malignancies (37). Among non-immunocompromised (18 cases, 30%), the main category was represented by patients with penetrating trauma (7/18, 39%). The most common sites of infection were sinus (35%) with/without CNS involvement, lung alone (25%), skin (20%), but in 11 cases (18%) dissemination was observed. According to EORTC criteria, the diagnosis of zygomycosis was proven in 46 patients (77%) and in most of them it was made in vivo (40/46 patients, 87%); in the remaining 14 cases (23%) the diagnosis was probable. 51 patients received antifungal therapy and in 30 of them surgical debridement was also performed. The most commonly used antifungal drug was liposomal amphotericin B (L-AmB), administered in 44 patients: 36 of these patients (82%) responded to therapy. Altogether an attributable mortality rate of 32% (19/60) was registered, which was reduced to 18% in patients treated with L-AmB (8/44). Zygomycosis is a rare and aggressive filamentous fungal infection, still associated with a high mortality rate. This study indicates an inversion of this trend, with a better prognosis and significantly lower mortality than that reported in the literature. It is possible that new extensive, aggressive diagnostic and therapeutic procedures, such as the use of L-AmB and surgery, have improved the prognosis of these patients.


Subject(s)
Zygomycosis/epidemiology , Adolescent , Adult , Aged , Aged, 80 and over , Child , Child, Preschool , Drug Resistance, Fungal , Female , Humans , Immunocompromised Host , Infant , Italy/epidemiology , Male , Middle Aged , Zygomycosis/diagnosis , Zygomycosis/drug therapy , Zygomycosis/etiology
3.
Dig Liver Dis ; 38(8): 609-11, 2006 Aug.
Article in English | MEDLINE | ID: mdl-16551516

ABSTRACT

Signet-ring cell carcinoma is a relatively rare neoplasm that rarely occurs in the urinary bladder. We report a case of a 60-year-old man who presented with gross haematuria. Cystoscopy revealed a white sessile tumour of the anterior bladder wall. The histological diagnosis showed a primary signet-ring cell carcinoma of the bladder (T3bN0M0). Eighteen months after radical cystectomy, the patient developed colon and stomach metastases. This case represents the first description of a primary signet-ring cell carcinoma of the urinary bladder with gastrointestinal metastases.


Subject(s)
Carcinoma, Signet Ring Cell/secondary , Colonic Neoplasms/secondary , Stomach Neoplasms/secondary , Urinary Bladder Neoplasms/pathology , Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Carcinoma, Signet Ring Cell/therapy , Colonic Neoplasms/surgery , Cystectomy , Cystoscopy , Fatal Outcome , Humans , Male , Middle Aged , Stomach Neoplasms/surgery , Urinary Bladder Neoplasms/therapy
4.
Proc Natl Acad Sci U S A ; 98(15): 8897-902, 2001 Jul 17.
Article in English | MEDLINE | ID: mdl-11447266

ABSTRACT

Portal hypertension resulting from increased intrahepatic resistance is a common complication of chronic liver diseases and a leading cause of death in patients with liver cirrhosis, a scarring process of the liver that includes components of both increased fibrogenesis and wound contraction. A reduced production of nitric oxide (NO) resulting from an impaired enzymatic function of endothelial NO synthase and an increased contraction of hepatic stellate cells (HSCs) have been demonstrated to contribute to high intrahepatic resistance in the cirrhotic liver. 2-(Acetyloxy) benzoic acid 3-(nitrooxymethyl) phenyl ester (NCX-1000) is a chemical entity obtained by adding an NO-releasing moiety to ursodeoxycholic acid (UDCA), a compound that is selectively metabolized by hepatocytes. In this study we have examined the effect of NCX-1000 and UDCA on liver fibrosis and portal hypertension induced by i.p. injection of carbon tetrachloride in rats. Our results demonstrated that although both treatments reduced liver collagen deposition, NCX-1000, but not UDCA, prevented ascite formation and reduced intrahepatic resistance in carbon tetrachloride-treated rats as measured by assessing portal perfusion pressure. In contrast to UDCA, NCX-1000 inhibited HSC contraction and exerted a relaxing effect similar to the NO donor S-nitroso-N-acetylpenicillamine. HSCs were able to metabolize NCX-1000 and release nitrite/nitrate in cell supernatants. In aggregate these data indicate that NCX-1000, releasing NO into the liver microcirculation, may provide a novel therapy for the treatment of patients with portal hypertension.


Subject(s)
Hypertension, Portal/prevention & control , Liver/metabolism , Nitrates , Nitric Oxide Donors/metabolism , Nitric Oxide/metabolism , Salicylates/metabolism , Ursodeoxycholic Acid/metabolism , Animals , Carbon Tetrachloride/pharmacology , Collagen/metabolism , Hypertension, Portal/metabolism , Liver/pathology , Liver Cirrhosis/physiopathology , Male , Nitric Oxide Donors/pharmacology , Rats , Rats, Wistar , Salicylates/pharmacology , Ursodeoxycholic Acid/pharmacology
5.
Gastroenterology ; 118(2): 404-21, 2000 Feb.
Article in English | MEDLINE | ID: mdl-10648469

ABSTRACT

BACKGROUND & AIMS: Concanavalin A (con A)-induced hepatitis is an immunomediated disease in which assembly of CD4(+) T cells and T helper (Th)1-like cytokines causes Fas-mediated liver cell death. Nitric oxide (NO) modulates Th1 response in vitro. NCX-4016 is an NO-aspirin derivative that spares the gastrointestinal tract and shares molecular targets with NO. The aim of this study was to investigate whether this NO-aspirin modulates Th1-like response induced by con A. METHODS: BALB/c mice were injected with 0.3 mg con A per mouse alone or in combination with NO-aspirin (18-100 mg/kg) or aspirin (10-55 mg/kg). RESULTS: NO-aspirin, but not aspirin, caused a dose-dependent protection against liver damage induced by con A. At a dose of 100 mg/kg, NO-aspirin caused a 40%-80% reduction of interleukin (IL)-1beta, IL-12, IL-18, interferon (IFN)-gamma, and tumor necrosis factor alpha production without affecting cytokine messenger RNA expression. NO-aspirin prevented Fas, Fas ligand, and IL-2 receptor up-regulation on spleen lymphocytes and Fas ligand on hepatocytes and caused the S-nitrosylation/inhibition of IL-1beta-converting enzyme-like cysteine proteases (caspases) involved in the processing and maturation of IL-1beta and IL-18. IL-18 immunoneutralization prevented IFN-gamma release and protected from liver injury induced by con A. In contrast to a selective caspase 1 inhibitor, zVAD.FMK, a pancaspase inhibitor, prevented IFN-gamma release and protected the liver from injury. CONCLUSIONS: Th1-like response induced by con A is mediated by IL-18 and requires activation of multiple caspases. NCX-4016 causes the S-nitrosylation/inhibition of caspases involved in cytokine production. Inhibition of Th1-like response is a new anti-inflammatory mechanism of action of NO-aspirin.


Subject(s)
Aspirin/analogs & derivatives , Caspases/metabolism , Chemical and Drug Induced Liver Injury/immunology , Concanavalin A/toxicity , Cytokines/immunology , Liver/pathology , Platelet Aggregation Inhibitors/pharmacology , T-Lymphocytes/immunology , Th1 Cells/immunology , Amino Acid Chloromethyl Ketones/pharmacology , Animals , Aspirin/pharmacology , Caspase 1/metabolism , Caspase 3 , Caspase Inhibitors , Chemical and Drug Induced Liver Injury/pathology , Chemical and Drug Induced Liver Injury/prevention & control , Cysteine Proteinase Inhibitors/pharmacology , Cytokines/biosynthesis , Cytokines/genetics , Fas Ligand Protein , Interferon-gamma/biosynthesis , Interleukin-18/physiology , Interleukins/biosynthesis , Liver/drug effects , Liver/immunology , Membrane Glycoproteins/genetics , Membrane Glycoproteins/physiology , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , RNA, Messenger/genetics , Receptors, Interleukin-2/genetics , Spleen/immunology , T-Lymphocytes/drug effects , Th1 Cells/drug effects , Transcription, Genetic/drug effects , Up-Regulation/drug effects , fas Receptor/genetics
6.
Ital J Gastroenterol Hepatol ; 31 Suppl 1: S6-13, 1999.
Article in English | MEDLINE | ID: mdl-10379463

ABSTRACT

Although non-steroidal anti-inflammatory drugs (NSAIDs) may impair the defensive ability of the gastric mucosal barrier through topical actions, recent evidence suggests that microcirculation disturbance plays a pivotal role in the genesis of gastric mucosal damage. In particular, attention has been drawn to the role exerted by those cytokine (TNF alpha and IL-1 beta) and adhesion molecules (LFA-1, Mac-1, ICAM-1) that regulate interactions between leukocyte and endothelial cells leading to gastric microvessels occlusion and ischaemic/hypoxic endothelial-epithelial cell damage. In recent years the role of prostaglandin synthesis inhibition in the pathogenesis of NSAID-gastropathy has been reconsidered, highlighting the immunomodulatory and pro-inflammatory consequences of prostanoids suppression. The awareness that mucosal damage is due to the non-discriminatory effect of NSAIDs on cyclo-oxygenase (COX) isoenzymes, has lead to the development of more selective, safer, COX-2 inhibitors. On the other hand, the observation that NO exerts a predominant physiological role in the maintenance of mucosal integrity has raised the opportunity to develop a new generation of NO-releasing NSAID derivatives with a reduced gastrointestinal toxicity.


Subject(s)
Anti-Inflammatory Agents, Non-Steroidal/adverse effects , Gastric Mucosa/drug effects , Gastric Mucosa/pathology , Stomach Ulcer/chemically induced , Stomach Ulcer/pathology , Biomarkers , Female , Humans , Intercellular Adhesion Molecule-1/drug effects , Intercellular Adhesion Molecule-1/metabolism , Leukotrienes/biosynthesis , Male , Prostaglandins/biosynthesis , Sensitivity and Specificity , Tumor Necrosis Factor-alpha/drug effects , Tumor Necrosis Factor-alpha/metabolism
7.
Gastroenterology ; 116(5): 1089-106, 1999 May.
Article in English | MEDLINE | ID: mdl-10220501

ABSTRACT

BACKGROUND & AIMS: Caspases, a class of cysteine proteases, modulate apoptosis. Nitric oxide (NO)-releasing nonsteroidal anti-inflammatory drugs (NSAIDs) are a new class of NSAID derivatives with reduced gastrointestinal toxicity. The aim of this study was to investigate whether cysteine endoproteases are involved in the pathogenesis of NSAID gastropathy and are target for NO-aspirin (NCX-4016). METHODS: Rats were treated orally with aspirin or equimolar doses of NCX-4016. Caspase activities were measured by fluorometric assay. Apoptosis was quantified by an enzyme-linked immunosorbent assay for histone-associated DNA, DNA ladder on agarose gel, and terminal deoxynucleotidyl transferase-mediated deoxyuridine triphosphate nick-end labeling assay. A primary culture of gastric chief cells was used to investigate whether NCX-4016 modulates guanosine 3',5'-cyclic monophosphate (cGMP)-dependent pathways. RESULTS: Short- and long-term (7 days) aspirin administration resulted in a time- and dose-dependent gastric injury that was associated with apoptosis and caspase up-regulation. Z-VAD.FMK, a pancaspase inhibitor, and NO donors protected from acute damage induced by aspirin. NCX-4016 spared the gastric mucosa and caused caspase inactivation by S-nitrosylation. Inhibition of tumor necrosis factor (TNF)-alpha release or activity by TAPI-2 or anti-TNF-alpha receptor monoclonal antibodies protected against mucosal damage and caspase activation. NCX-4016 protected gastric chief cells from toxicity induced by TNF-alpha by activating cGMP-dependent pathways. CONCLUSIONS: Aspirin administration leads to a TNF-alpha-dependent activation of gastric caspases. NO-aspirin spares the gastric mucosa and inhibits caspase activity through cGMP-dependent and -independent pathways.


Subject(s)
Anti-Inflammatory Agents, Non-Steroidal/pharmacology , Aspirin/analogs & derivatives , Caspases/metabolism , Animals , Anti-Inflammatory Agents, Non-Steroidal/administration & dosage , Anti-Inflammatory Agents, Non-Steroidal/adverse effects , Apoptosis , Aspirin/administration & dosage , Aspirin/adverse effects , Aspirin/metabolism , Aspirin/pharmacology , Caspase Inhibitors , Cyclic GMP/metabolism , Cysteine Proteinase Inhibitors/pharmacology , Dose-Response Relationship, Drug , Gastric Mucosa/cytology , Gastric Mucosa/drug effects , Gastric Mucosa/enzymology , Gastric Mucosa/injuries , Humans , In Situ Nick-End Labeling , Nitric Oxide/metabolism , Nitric Oxide Donors/pharmacology , Nitroprusside/pharmacology , Rats , Salicylates/blood , Time Factors , Tumor Necrosis Factor-alpha/metabolism , Tumor Necrosis Factor-alpha/pharmacology , U937 Cells
8.
Aliment Pharmacol Ther ; 13(3): 421-35, 1999 Mar.
Article in English | MEDLINE | ID: mdl-10102977

ABSTRACT

BACKGROUND: Nitric oxide (NO)-releasing NSAIDs are a new class of NSAID derivatives with markedly reduced gastrointestinal toxicity. Although it has been demonstrated that NO-NSAIDs spare gastric mucosal blood flow, molecular determinants involved in this effect are unknown. AIM: To investigate the effect of aspirin, naproxen and flurbiprofen, and their NO-derivatives, on gastric apoptosis and endothelial cell damage induced by tumour necrosis factor-alpha (TNFalpha). In other systems, TNFalpha-induced apoptosis is mediated by caspases, a growing family of cysteine proteases similar to the IL-1beta converting enzyme (ICE), and so we have investigated whether NO-NSAIDs modulate ICE-like endopeptidases. METHODS: Rats were treated orally with aspirin, naproxen and flurbiprofen, or their NO-releasing derivatives in equimolar doses, and were killed 3 h later to assess mucosal damage and caspase activity. Endothelial cells (HUVECs) were obtained from human umbilical cord by enzymatic digestion. Caspase 1 and 3 activities were measured by a fluorimetric assay using selective peptides as substrates and inhibitors. Apoptosis was quantified by ELISA specific for histone-associated DNA fragments and by the terminal transferase nick-end translation method (TUNEL). RESULTS: In vivo NSAID administration caused a time-dependent increase in gastric mucosal damage and caspase activity. NCX-4016, NO-naproxen and NO-flurbiprofen did not cause any mucosal damage and prevented cysteine protease activation. NSAIDs and NO-NSAIDs stimulated TNFalpha release. Exposure to TNFalpha resulted in a time- and concentration-dependent HUVEC apoptosis, an effect that was prevented by pretreating the cells with NCX-4016, NO-naproxen, NO-flurbiprofen, SNP or Z-VAD.FMK, a pan-caspase inhibitor. The activation of ICE-like cysteine proteases was required to mediate TNFalpha-induced apoptosis of HUVECs. Exogenous NO donors inhibited TNFalpha-induced cysteine protease activation. Inhibition of caspase activity was due to S-nitrosylation of ICE/CPP32-like proteases. NO-NSAIDs prevented IL-1beta release from endotoxin-stimulated macrophages. CONCLUSIONS: NO-releasing NSAIDs are a new class of non-peptide caspase inhibitors. Inhibition of ICE-like cysteine proteases prevents endothelial cell damage induced by pro-inflammatory agents and might contribute to the gastro-protective effects of NO-NSAIDs.


Subject(s)
Anti-Inflammatory Agents, Non-Steroidal/pharmacology , Apoptosis/drug effects , Caspase Inhibitors , Cysteine Proteinase Inhibitors/pharmacology , Gastric Mucosa/cytology , Nitric Oxide/metabolism , Animals , Aspirin/analogs & derivatives , Aspirin/pharmacology , Cell Line , DNA Fragmentation/drug effects , Endothelium, Vascular/cytology , Endothelium, Vascular/drug effects , Flurbiprofen/analogs & derivatives , Flurbiprofen/pharmacology , Gastric Mucosa/drug effects , Gastric Mucosa/enzymology , Humans , In Situ Nick-End Labeling , Male , Mice , Naproxen/analogs & derivatives , Naproxen/pharmacology , Nitric Oxide/pharmacology , Peroxidase/antagonists & inhibitors , Peroxidase/metabolism , Rats , Rats, Wistar
9.
Aliment Pharmacol Ther ; 12(11): 1139-53, 1998 Nov.
Article in English | MEDLINE | ID: mdl-9845404

ABSTRACT

BACKGROUND: Although previous studies indicate that prevention of tumour necrosis factor alpha (TNFalpha) release protects against NSAID-induced gastric mucosal injury, intracellular pathways by which aspirin causes TNFalpha release are unknown. TNFalpha is synthesized as a precursor which is proteolytically cleaved by a specific converting enzyme, TACE, to release the mature cytokine. TACE inhibitors prevent TNFalpha release and protect against TNFalpha-mediated disease. AIM: To investigate: (i) molecular events that regulate TNFalpha secretion in response to aspirin in vivo and in vitro; (ii) whether TNFalpha secretion inhibitors prevent aspirin-induced TNFalpha release and protect against gastric mucosal damage; and (iii) whether TNFalpha exerts a direct cytotoxic effect on gastric epithelial cells. METHODS: In vitro studies were carried out on mouse macrophages and rat gastric mucosal cells. Gastric mucosal damage was induced in rats by oral administration of 300 mg/kg aspirin. TNFalpha cytotoxicity on gastric mucosal cells was examined by treating rats with lipopolysaccharide to release TNFalpha or by incubating dispersed gastric mucosal cells with increasing concentrations of TNFalpha. RESULTS: Aspirin increases intracellular calcium (Ca2+) levels and causes a time and concentration dependent increase in macrophage TNFalpha mRNA accumulation and cytokine release. Agents that cause Ca2+ mobilization with a receptor-independent mechanism, such as ionomycin and thapsigargin, stimulate TNFalpha release. Incubating the macrophages in a Ca2+ free medium inhibited TNFalpha secretion. Agents that prevent TNFalpha mRNA transcription, e.g. lisophylline, PGE2, interleukin-10 and 8-BrcAMP, or TACE inhibitors, e.g. EDTA, TAPI-2 and BB-3103, inhibit TNFalpha release and protect rats against gastric mucosal injury induced by oral administration of aspirin. TNFalpha exerts a direct cytotoxic effect on gastric epithelial cells as demonstrated by the reduced viability observed in gastric mucosal cells prepared from rats treated with lipopolysaccharide, or directly incubated with increasing concentrations of TNFalpha. CONCLUSIONS: (i) Aspirin directly stimulates TNFalpha gene transcription; (ii) TACE inhibitors protect against aspirin-induced gastric mucosal injury; and (iii) TNFalpha exerts a direct cytotoxic effect on gastric epithelial cells.


Subject(s)
Aspirin/pharmacology , Gastric Mucosa/drug effects , Tumor Necrosis Factor-alpha/metabolism , Animals , Anti-Inflammatory Agents, Non-Steroidal/pharmacology , Blotting, Northern , Cells, Cultured , Edetic Acid/pharmacology , Hydroxamic Acids/pharmacology , Macrophages/metabolism , Male , Mice , Pentoxifylline/analogs & derivatives , Pentoxifylline/pharmacology , RNA, Messenger/metabolism , Rats , Rats, Wistar
10.
Ital J Gastroenterol Hepatol ; 30(4): 385-90, 1998 Aug.
Article in English | MEDLINE | ID: mdl-9789134

ABSTRACT

BACKGROUND AND AIMS: A new amino acid derivative of 5-aminosalicylic acid, 5-ASA-glutamate, releases 5-aminosalicylic acid independently of the action of bacterial azoreductases or adapt intestinal pH. In this study, 5-ASA-glutamate was compared with sulphasalazine with respect to: 1) therapeutic action, 2) effects on the synthesis of eicosanoids, 3) regional release of 5-aminosalicylic acid in the intestine. METHODS: Colitis was induced in 29 rats by intracolonic administration of trinitrobenzensulfonic acid. Nine animals received an equal amount of saline. Three days after induction of colitis, animals were randomly assigned to equimolecular doses of 5-aminosalicylic acid as sulphasalazine (1040 mg/kg bw day) or 5-ASA-glutamate (850 mg/kg bw day) or arabic gum in water, given intragastrically. Arabic gum was also administered to animals that had received a saline enema (control group). The guts of 3 rats from the 5-ASA-glutamate group and 3 from the sulphasalazine group were used to assess regional release of 5-ASA, while in all the others, after 21 days of treatment, macroscopic and histologic lesions were assessed and eicosanoids and leukotriene determinations were performed. RESULTS: The 5-ASA-glutamate group had macroscopic (2.20 +/- 0.58) and histologic (2.80 +/- 1.24) significantly lower scores than the trinitrobenzensulfonic acid group (3.40 +/- 0.22 and 6.50 +/- 1.2 respectively). 5-ASA-glutamate group had reduced PGE2 (-31%) and TXB2 (-25%) more effectively than the sulphasalazine group. LTB4 release was not affected by 5-ASA-glutamate treatment, while sulphasalazine produced a non significant, but quite consistent, reduction in LTB4 release (-37%). The release of 5-ASA after sulphasalazine was higher in the small intestine, lower in the colon compared to that following 5-ASA-glutamate administration. CONCLUSIONS: 5ASA-glutamate was effective in reducing the macroscopic and histologic score in the trinitrobenzensulfonic acid induced colitis. It also had some effect in reducing eicosanoid synthesis and could be a promising drug for the treatment of inflammatory bowel disease.


Subject(s)
Colitis/drug therapy , Glutamates/therapeutic use , Animals , Anti-Inflammatory Agents, Non-Steroidal/pharmacology , Anti-Inflammatory Agents, Non-Steroidal/therapeutic use , Chromatography, High Pressure Liquid , Colitis/chemically induced , Colitis/metabolism , Colon/drug effects , Colon/pathology , Disease Models, Animal , Eicosanoids/biosynthesis , Glutamates/pharmacokinetics , Leukotrienes/biosynthesis , Prostaglandins/biosynthesis , Radioimmunoassay , Random Allocation , Rats , Rats, Sprague-Dawley , Statistics, Nonparametric , Sulfasalazine/pharmacology , Sulfasalazine/therapeutic use , Trinitrobenzenesulfonic Acid
12.
Kidney Int ; 32(5): 754-9, 1987 Nov.
Article in English | MEDLINE | ID: mdl-2448515

ABSTRACT

Red blood cell (RBC) sodium transport systems were studied by cation flux methodology, measuring both the ouabain-sensitive Na-K pump and the ouabain-insensitive Na-K cotransport (CoT), as well as the Na-lithium (Li) countertransport (CTT), in eight patients on chronic hemodialysis and a control group of eight normal individuals. Intracellular sodium content and passive Na permeability were also determined. The effect of L-carnitine on RBC sodium transport in the uremic group was evaluated by supplementation with oral (1 g/day) and i.v. (1 g post-hemodialysis) L-carnitine for 60 days. Mean Na efflux through the ouabain-sensitive Na-K pump was 30.7% lower in uremic patients than in controls (3.49 +/- 1.52 vs. 5.04 +/- 0.72 mmol/liter RBCxhr; P less than 0.025). Intracellular Na content was higher in uremic patients (11.57 +/- 3.38 vs. 8.86 +/- 0.88 mEq/liter RBC; P less than 0.05), but no differences were found in Na-K CoT, Na-Li CTT or passive Na permeability. L-carnitine treatment increased the ouabain-sensitive Na efflux in uremic patients (4.76 +/- 1.6 vs. 3.49 +/- 1.52 mmol/liter RBCxhr; P less than 0.05), with no change in CoT, CTT, intracellular Na content or passive Na permeability. We conclude that L-carnitine deficiency may play a major role in uremic Na-K pump disfunction.


Subject(s)
Carnitine/administration & dosage , Erythrocytes/metabolism , Renal Dialysis , Sodium/blood , Uremia/blood , Administration, Oral , Adult , Cell Membrane Permeability/drug effects , Female , Humans , Ion Channels/drug effects , Lithium/blood , Male , Middle Aged , Potassium/blood
SELECTION OF CITATIONS
SEARCH DETAIL
...