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

Publication year range
1.
Haemophilia ; 23(5): 673-681, 2017 Sep.
Article in English | MEDLINE | ID: mdl-28594466

ABSTRACT

Bleeding diathesis has been considered for a long time the main clinical issue impacting the lives of patients affected by inherited thrombocytopaenias. However, the number of known inherited thrombocytopaenias greatly increased in recent years, and careful evaluation of hundreds of patients affected by these 'new' disorders revealed that most of them are at risk of developing additional life-threatening disorders during childhood or adult life. These additional disorders are usually more serious and dangerous than low platelet count. For instance, it is known that mutations in RUNX1, ANKRD26 and ETV6 cause congenital thrombocytopaenia, but we now know that they also predispose to haematological malignancies. Similarly, MYH9 mutations result in congenital thrombocytopaenia and increase the risk of developing kidney failure, cataracts and hearing loss at a later stage, while MPL mutations cause a congenital thrombocytopaenia that almost always evolves into deadly bone marrow failure. Thus, identification of patients with these disorders is essential for evaluation of their prognosis, enabling effective genetic counselling, personalizing follow-up and giving appropriate treatments in case of development of additional diseases. Careful clinical evaluation and peripheral blood film examination are extremely useful tools in guiding the diagnostic process and identifying the candidate genes to be sequenced.


Subject(s)
Genetic Diseases, Inborn/complications , Hemorrhage/etiology , Thrombocytopenia/complications , Clinical Decision-Making , Diagnosis, Differential , Disease Management , Genetic Association Studies , Genetic Diseases, Inborn/diagnosis , Genetic Diseases, Inborn/therapy , Genetic Predisposition to Disease , Hemorrhage/diagnosis , Hemorrhage/therapy , Humans , Phenotype , Thrombocytopenia/diagnosis , Thrombocytopenia/therapy
2.
Nat Genet ; 26(1): 103-5, 2000 Sep.
Article in English | MEDLINE | ID: mdl-10973259

ABSTRACT

The autosomal dominant, giant-platelet disorders, May-Hegglin anomaly (MHA; MIM 155100), Fechtner syndrome (FTNS; MIM 153640) and Sebastian syndrome (SBS), share the triad of thrombocytopenia, large platelets and characteristic leukocyte inclusions ('Döhle-like' bodies). MHA and SBS can be differentiated by subtle ultrastructural leukocyte inclusion features, whereas FTNS is distinguished by the additional Alport-like clinical features of sensorineural deafness, cataracts and nephritis. The similarities between these platelet disorders and our recent refinement of the MHA (ref. 6) and FTNS (ref. 7) disease loci to an overlapping region of 480 kb on chromosome 22 suggested that all three disorders are allelic. Among the identified candidate genes is the gene encoding nonmuscle myosin heavy chain 9 (MYH9; refs 8-10), which is expressed in platelets and upregulated during granulocyte differentiation. We identified six MYH9 mutations (one nonsense and five missense) in seven unrelated probands from MHA, SBS and FTNS families. On the basis of molecular modelling, the two mutations affecting the myosin head were predicted to impose electrostatic and conformational changes, whereas the truncating mutation deleted the unique carboxy-terminal tailpiece. The remaining missense mutations, all affecting highly conserved coiled-coil domain positions, imparted destabilizing electrostatic and polar changes. Thus, our results suggest that mutations in MYH9 result in three megakaryocyte/platelet/leukocyte syndromes and are important in the pathogenesis of sensorineural deafness, cataracts and nephritis.


Subject(s)
Blood Platelet Disorders/genetics , Leukocytes/pathology , Molecular Motor Proteins , Mutation , Myosin Heavy Chains/genetics , Alleles , Amino Acid Sequence , Animals , Blood Platelet Disorders/pathology , Cataract/genetics , Chickens , Chromosomes, Human, Pair 22 , Crystallography, X-Ray , Cytoplasm/metabolism , Genotype , Hearing Loss, Sensorineural/genetics , Humans , Models, Molecular , Molecular Sequence Data , Muscle, Smooth/metabolism , Mutation, Missense , Myosin Heavy Chains/chemistry , Myosins/chemistry , Myosins/genetics , Nephritis/genetics , Neutrophils/pathology , Neutrophils/ultrastructure , Phenotype , Protein Conformation , Protein Structure, Tertiary , Sequence Homology, Amino Acid , Syndrome , Thrombocytopenia/genetics
3.
J Thromb Haemost ; 16(9): 1700-1710, 2018 09.
Article in English | MEDLINE | ID: mdl-29956472

ABSTRACT

Hereditary thrombocytopenias (HTPs) constitute a heterogeneous group of diseases characterized by a reduction in platelet count and a potential bleeding risk. As a result of advances in diagnostic methods, HTPs are increasingly being identified, and appear to be less rare than previously thought. Most HTPs do not have effective treatments, except for platelet transfusion when bleeding occurs and in preparation for procedures associated with a risk of bleeding. Preliminary clinical evidence suggests that thrombopoietin receptor agonists (TPO-RAs) with an established use in the treatment of certain acquired thrombocytopenias are well tolerated and provide clinical benefits in patients with some forms of HTP. These drugs may therefore be considered for the treatment of HTPs in clinical practice. However, caution and close monitoring are recommended, owing to the absence of long-term safety data and the potential risks posed by prolonged bone marrow stimulation in certain HTPs. In this review, we summarize the available clinical data on TPO-RAs in the treatment of HTPs, and discuss their use in patients with these disorders. We believe that TPO-RAs will play a major role in the treatment of HTPs, particularly myosin heavy chain 9-related disease, Wiskott-Aldrich syndrome, X-linked thrombocytopenia, and thrombocytopenia caused by THPO mutations.


Subject(s)
Benzoates/therapeutic use , Hydrazines/therapeutic use , Pyrazoles/therapeutic use , Receptors, Fc/therapeutic use , Receptors, Thrombopoietin/agonists , Recombinant Fusion Proteins/therapeutic use , Thrombocytopenia/drug therapy , Thrombopoietin/therapeutic use , Benzoates/adverse effects , Benzoates/pharmacology , Clinical Trials, Phase I as Topic , Clinical Trials, Phase II as Topic , Genetic Association Studies , Genetic Heterogeneity , Genetic Predisposition to Disease , Hematologic Neoplasms/etiology , Humans , Hydrazines/adverse effects , Hydrazines/pharmacology , Primary Myelofibrosis/etiology , Purpura, Thrombocytopenic, Idiopathic/complications , Purpura, Thrombocytopenic, Idiopathic/drug therapy , Pyrazoles/adverse effects , Pyrazoles/pharmacology , Recombinant Fusion Proteins/adverse effects , Recombinant Fusion Proteins/pharmacology , Risk , Thrombocytopenia/genetics , Thrombophilia/chemically induced , Thrombopoiesis/drug effects , Thrombopoietin/adverse effects , Thrombopoietin/pharmacology
4.
J Thromb Haemost ; 5(3): 551-9, 2007 Mar.
Article in English | MEDLINE | ID: mdl-17137471

ABSTRACT

BACKGROUND: We report a novel case of gray platelet syndrome (GPS). A 14-year-old boy had bleeding diathesis, mild thrombocytopenia, giant platelets with severe defect of alpha-granule secretory proteins, myelofibrosis and splenomegaly. METHODS AND RESULTS: Platelet function studies showed a marked reduction of aggregation and Ca(2+) mobilization by thrombin, protease-activated receptor 1 (PAR1)-activating peptide (AP) and PAR4-AP, PAR1 expression at 55% of normal levels, and a more than two hundred fold reduction of in vitro whole-blood thromboxane B(2) (TXB(2)) production. Sequencing of coding regions of the PAR1 gene failed to show abnormalities. This patient was initially classified as a sporadic case of GPS, as electron microscopy failed to identify giant platelets and/or alpha-granule deficiency in his relatives. However, further studies on the father and three other relatives showed a relative lack of platelet alpha-granule proteins by immunofluorescence microscopy, a defective platelet response to PAR4-AP, and severely reduced in vitro whole-blood TXB(2) production. On this basis, we suggest that in this family, GPS was transmitted in a dominant fashion with highly variable penetrance. CONCLUSIONS: Our study suggests that current diagnostic criteria fail to identify some patients with a mild GPS phenotype and that such patients might be identified by the methods cited above. It also better characterizes the pathogenesis of defective platelet responses to thrombin, and raises interesting questions on the correlation between abnormal PAR function and the lack of alpha-granule content in GPS.


Subject(s)
Blood Platelets/drug effects , Coagulants/pharmacology , Platelet Aggregation/drug effects , Platelet Storage Pool Deficiency/blood , Receptor, PAR-1/agonists , Thrombin/pharmacology , Adolescent , Adult , Aged , Blood Platelets/metabolism , Blood Platelets/ultrastructure , Calcium Signaling/drug effects , Cytoplasmic Granules/ultrastructure , Family , Female , Humans , Male , Microscopy, Fluorescence , Middle Aged , Oligopeptides/pharmacology , P-Selectin/analysis , Pedigree , Phenotype , Platelet Factor 4/analysis , Platelet Function Tests , Platelet Storage Pool Deficiency/diagnosis , Platelet Storage Pool Deficiency/genetics , Platelet Storage Pool Deficiency/metabolism , Platelet Storage Pool Deficiency/pathology , Receptor, PAR-1/genetics , Receptor, PAR-1/metabolism , Syndrome , Thrombospondin 1/analysis , Thromboxane B2/blood
5.
J Thromb Haemost ; 15(11): 2226-2229, 2017 11.
Article in English | MEDLINE | ID: mdl-28921865

ABSTRACT

Essentials Extramedullary hematopoiesis (EMH) represents a pathologic finding in adult life. We report a mass-like EMH in the presacral space in a patient with ANKRD26-related thrombocytopenia. We found possible correlation between EMH and conditions causing lifelong thrombocytopenia. EMH can cause masses of unknown origin in patients with inherited thrombocytopenias. SUMMARY: Most commonly located in the liver and spleen, extramedullary hematopoiesis (EMH) is the presence of hematopoietic tissue outside the bone marrow. MYH9-related thrombocytopenia (MYH9-RD) and ANKRD26-related thrombocytopenia (ANKRD26-RT) are two of the most frequent forms of inherited thrombocytopenia (IT). Until recently, EMH has been associated with neoplastic and non-neoplastic hematologic conditions in which ITs were not included. We describe a case of mass-like EMH in the presacral space in a patient affected with ANKRD26-RT, comparing it with another case of paravertebral EMH we recently described in a subject with MYH9-RD. The surprisingly similitude of such a finding in the context of a group of rare disorders induces us to speculate about the possible pathogenic relationship between EMH and conditions causing lifelong thrombocytopenia, particularly the entity of ITs. Finally, we suggest that EMH has to be taken into consideration in the diagnostic work-up of masses of unknown origin in subjects affected with ITs.


Subject(s)
Hematopoiesis, Extramedullary/genetics , Mutation , Nuclear Proteins/genetics , Thrombocytopenia/genetics , Aged , Biopsy, Needle , Bone Marrow Examination , DNA Mutational Analysis , Female , Genetic Predisposition to Disease , Heredity , Humans , Immunohistochemistry , Intercellular Signaling Peptides and Proteins , Pelvis , Phenotype , Positron Emission Tomography Computed Tomography , Spleen/diagnostic imaging , Thrombocytopenia/blood , Thrombocytopenia/diagnosis
6.
J Thromb Haemost ; 15(7): 1511-1521, 2017 07.
Article in English | MEDLINE | ID: mdl-28457011

ABSTRACT

Essentials There are many hereditary platelet disorders (HPD) but diagnosing these is challenging. We provide a method to diagnose several HPDs using standard blood smears requiring < 100 µL blood. By this approach, the underlying cause of HPD was characterized in ~25-30% of referred individuals. The method facilitates diagnosis of HPD for patients of all ages around the world. SUMMARY: Background Many hereditary thrombocytopenias and/or platelet function disorders have been identified, but diagnosis of these conditions remains challenging. Diagnostic laboratory techniques are available only in a few specialized centers and, using fresh blood, often require the patient to travel long distances. For the same reasons, patients living in developing countries usually have limited access to diagnosis. Further, the required amount of blood is often prohibitive for pediatric patients. Objectives By a collaborative international approach of four centers, we aimed to overcome these limitations by developing a method using blood smears prepared from less than 100 µL blood, for a systematic diagnostic approach to characterize the platelet phenotype. Methods We applied immunofluorescence labelling (performed centrally) to standard air-dried peripheral blood smears (prepared locally, shipped by regular mail), using antibodies specific for proteins known to be affected in specific hereditary platelet disorders. Results By immunofluorescence labelling of blood smears we characterized the underlying cause in 877/3217 (27%) patients with suspected hereditary platelet disorders (HPD). Currently about 50 genetic causes for HPD are identified. Among those, the blood smear method was especially helpful to identify MYH9 disorders/MYH9-related disease, biallelic Bernard-Soulier syndrome, Glanzmann thrombasthenia and gray platelet syndrome. Diagnosis could be established for GATA1 macrothrombocytopenia, GFI1B macrothrombocytopenia, ß1-tubulin macrothrombocytopenia, filamin A-related thrombocytopenia and Wiskott-Aldrich syndrome. Conclusion Combining basic and widely available preanalytical methods with the immunomorphological techniques presented here, allows detailed characterization of the platelet phenotype. This supports genetic testing and facilitates diagnosis of hereditary platelet disorders for patients of all ages around the world.


Subject(s)
Blood Platelet Disorders/blood , Blood Platelet Disorders/diagnosis , Blood Platelets/metabolism , Hematologic Tests/instrumentation , Hematologic Tests/methods , Alleles , Bernard-Soulier Syndrome/genetics , Female , Humans , Immunophenotyping , International Cooperation , Male , Microscopy, Fluorescence , Molecular Motor Proteins/genetics , Myosin Heavy Chains/genetics , Phenotype , Sensitivity and Specificity , Thrombasthenia/genetics
7.
J Thromb Haemost ; 4(4): 848-60, 2006 Apr.
Article in English | MEDLINE | ID: mdl-16634756

ABSTRACT

BACKGROUND: Megakaryopoiesis represents a multi-step, often unclear, process leading to commitment, differentiation, and maturation of megakaryocytes (MKs) that release platelets. AIM: To identify the novel genes that might help to clarify the molecular mechanisms of megakaryocytopoiesis and be regarded as potential candidates of inherited platelet defects, global gene expression of hematopoietic lineages was carried out. METHODS: Human cord blood was used to purify CD34+ stem cells and in vitro expand CD41+ cells and burst-forming unit-erythroid (BFU-E). We investigated the expression profiles of these three hematopoietic lineages in the Affymetrix system and selected genes specifically expressed in MKs by comparing transcripts of the different lineages using the dchip and pam algorithms. RESULTS: A detailed characterization of MK population showed that 99% of cells expressed the CD41 antigen whereas 73% were recognizable as terminally differentiated fetal MKs. The profile of these cells was compared with that of CD34+ cells and BFU-E allowing us to select 70 transcripts (MK-core), which represent not only the genes with a well-known function in MKs, but also novel genes never detected or characterized in these cells. Moreover, the specific expression was confirmed at both RNA and protein levels, thus validating the 'MK-core' isolated by informatics tools. CONCLUSIONS: This is a global gene expression that for the first time depicts a well-characterized population of cord blood-derived fetal MKs. Novel genes have been detected, such as those encoding components of the extracellular matrix and basal membrane, which have been found in the cytoplasm of Mks, suggesting that new physiological aspects of MKs should be studied.


Subject(s)
Fetal Blood/cytology , Platelet Membrane Glycoprotein IIb/biosynthesis , Thrombopoiesis/physiology , Antibodies, Monoclonal/metabolism , Antigens, CD34/biosynthesis , Antigens, CD34/metabolism , Erythroid Precursor Cells/metabolism , Flow Cytometry , Humans , In Vitro Techniques , Microscopy, Fluorescence , Multigene Family , Oligonucleotide Array Sequence Analysis , Platelet Membrane Glycoprotein IIb/chemistry , RNA/chemistry , RNA/metabolism , Reverse Transcriptase Polymerase Chain Reaction
8.
J Thromb Haemost ; 3(5): 1026-35, 2005 May.
Article in English | MEDLINE | ID: mdl-15869600

ABSTRACT

MYH9-related disease (MYH9-RD) is an autosomal dominant disorder deriving from mutations in the MYH9 gene encoding for the heavy chain of non-muscle myosin IIA, and characterized by thrombocytopenia and giant platelets. Isoform IIA of myosin is the only one expressed in platelets, but the possibility that MYH9 mutations affect the organization of contractile structures in these blood elements has never been investigated. In this work we have analyzed the composition and the agonist-induced reorganization of the platelet cytoskeleton from seven MYH9-RD patients belonging to four different families. We found that an increased amount of myosin was constitutively associated with actin in the cytoskeleton of resting MYH9-RD platelets. Upon platelet stimulation, an impaired increase in the total cytoskeletal proteins was observed. Moreover, selected membrane glycoproteins, tyrosine kinases, and small GTPases failed to interact with the cytoskeleton in agonist-stimulated MYH9-RD platelets. These results demonstrate for the first time that mutations of MYH9 result in an alteration of the composition and agonist-induced reorganization of the platelet cytoskeleton. We suggest that these abnormalities may represent the biochemical basis for the previously reported functional alterations of MYH9-RD platelets, and for the abnormal platelet formation from megakaryocytes, resulting in thrombocytopenia and giant platelets.


Subject(s)
Blood Platelet Disorders/diagnosis , Blood Platelet Disorders/metabolism , Blood Platelets/metabolism , Cytoskeleton/metabolism , Molecular Motor Proteins/metabolism , Molecular Motor Proteins/physiology , Myosin Heavy Chains/metabolism , Myosin Heavy Chains/physiology , Thrombocytopenia/genetics , Adolescent , Adult , Dimerization , Electrophoresis, Polyacrylamide Gel , Family Health , Female , GTP Phosphohydrolases/metabolism , Genes, Dominant , Glycoproteins/metabolism , Humans , Immunoblotting , Male , Megakaryocytes/metabolism , Middle Aged , Mutation , Nonmuscle Myosin Type IIA/chemistry , Polymorphism, Genetic , Signal Transduction
9.
Leukemia ; 15(12): 1885-91, 2001 Dec.
Article in English | MEDLINE | ID: mdl-11753609

ABSTRACT

Patients undergoing allogeneic hematopoietic stem cell transplantation (HSCT) always require platelet transfusions, but the increase in platelet count is often less than expected. Since factors responsible for poor response to platelet transfusions in this clinical setting are largely unknown, we performed a prospective study in 87 consecutive children transplanted in a single institution. The mean 16-h corrected count increment (CCI) of 598 platelet transfusions was 5.76 +/- 8.32 x 10(9)/l. Both before and after HSCT, 13.8% of patients had antibodies against HLA and/or platelet-specific antigens. Univariate analysis identified 12 factors significantly associated with a lower post-transfusion CCI, but only four reached statistical significance in the multivariate analysis. These four factors were concomitant therapy with vancomycin, alloimmunization, use of an Autopheresis cell separator for preparation of platelet concentrates and cytomegalovirus infection. We, therefore, suggest that a better response to platelet transfusions could be obtained by choosing a suitable cell separator, by avoiding the use of vancomycin and by adopting measures that reduce alloimmunization and CMV infection. Moreover, screening patients for HLA and platelet-specific antibodies before HSCT would identify the majority of subjects who will develop alloimmune refractoriness after transplantation and would allow the search for a compatible donor in advance.


Subject(s)
Hematopoietic Stem Cell Transplantation , Platelet Transfusion/standards , Analysis of Variance , Anti-Bacterial Agents/adverse effects , Anti-Bacterial Agents/therapeutic use , Antigens, Human Platelet/immunology , Child , Child, Preschool , Contraindications , Cytapheresis/instrumentation , Cytomegalovirus Infections/blood , Cytomegalovirus Infections/complications , Female , HLA Antigens/immunology , Hematologic Diseases/therapy , Humans , Infant , Isoantibodies/blood , Male , Platelet Count , Prospective Studies , Transplantation Immunology , Transplantation, Homologous/immunology , Vancomycin/adverse effects , Vancomycin/therapeutic use
10.
Exp Hematol ; 14(5): 338-42, 1986 Jun.
Article in English | MEDLINE | ID: mdl-3011483

ABSTRACT

A murine monoclonal antibody against glycoproteins IIb-IIIa of platelet membrane completely abolished platelet aggregation induced by epinephrine, arachidonic acid, and low concentration of collagen and thrombin, but it had only minor inhibitory effects on aggregation induced by ADP, higher amounts of collagen and thrombin, and these agents combined in pairs. The simultaneous studies of aggregation and ATP secretion demonstrated that aggregation plays an essential role in stimulating the platelet-release reaction, except for the thrombin-induced ATP secretion that seems to be largely dependent on fibrinogen binding to platelet surface.


Subject(s)
Adenosine Triphosphate/metabolism , Antibodies, Monoclonal/pharmacology , Platelet Aggregation , Receptors, Cell Surface/immunology , Animals , Arachidonic Acid , Arachidonic Acids/pharmacology , Collagen/pharmacology , Epinephrine/pharmacology , Humans , Mice , Platelet Membrane Glycoproteins , Thrombin/pharmacology
11.
J Thromb Haemost ; 13(4): 651-9, 2015 Apr.
Article in English | MEDLINE | ID: mdl-25529050

ABSTRACT

BACKGROUND: Single nucleotide polymorphisms (SNPs) in platelet-associated genes partly explain inherent variability in platelet counts. Patients with monoallelic Bernard Soulier syndrome due to the Bolzano mutation (GPIBA A156V) have variable platelet counts despite a common mutation for unknown reasons. OBJECTIVES: We investigated the effect of the most common SNP (R307H) in the hematopoietic-specific tubulin isotype ß-1 in these Bernard Soulier patients and potential microtubule-based mechanisms of worsened thrombocytopenia. PATIENTS/METHODS: Ninety-four monoallelic Bolzano mutation patients were evaluated for the R307H ß-1 SNP and had platelet counts measured by three methods; the Q43P SNP was also evaluated. To investigate possible mechanisms underlying this association, we used molecular modeling of ß-1 tubulin with and without the R307H SNP. We transfected SNP or non-SNP ß-1 tubulin into MCF-7 and CMK cell lines and measured microtubule regrowth after nocodazole-induced depolymerization. RESULTS: We found that patients with at least one R307H SNP allele had significantly worse thrombocytopenia; manual platelet counting revealed a median platelet count of 124 in non-SNP patients and 76 in SNP patients (both ×10(9)  L(-1) ; P < 0.01). The Q43P SNP had no significant association with platelet count. Molecular modeling suggested a structural relationship between the R307H SNP and microtubule stability via alterations in the M-loop of ß tubulin; in vitro microtubule recovery assays revealed that cells transfected with R307H SNP ß-1 had significantly impaired microtubule recovery. CONCLUSIONS: Our data show that the R307H SNP is significantly associated with the degree of thrombocytopenia in congenital and acquired platelet disorders, and may affect platelets by altering microtubule behavior.


Subject(s)
Bernard-Soulier Syndrome/genetics , Blood Platelets/metabolism , Microtubules/metabolism , Polymorphism, Single Nucleotide , Tubulin/genetics , Tubulin/metabolism , Bernard-Soulier Syndrome/blood , Bernard-Soulier Syndrome/diagnosis , Blood Platelets/drug effects , Crystallography, X-Ray , Genetic Markers , Genetic Predisposition to Disease , Humans , MCF-7 Cells , Microtubules/drug effects , Models, Molecular , Phenotype , Platelet Count , Protein Conformation , Protein Stability , Severity of Illness Index , Structure-Activity Relationship , Transfection , Tubulin/chemistry , Tubulin Modulators/pharmacology
12.
Am J Med ; 104(4): 355-60, 1998 Apr.
Article in English | MEDLINE | ID: mdl-9576409

ABSTRACT

PURPOSE: May-Hegglin anomaly is a rare hereditary condition characterized by the triad of thrombocytopenia, giant platelets, and inclusion bodies in leukocytes. Clinical features and the pathogenesis of bleeding in this disease are poorly defined. PATIENTS AND METHODS: From 1988 to 1996 we studied 15 new May-Hegglin anomaly patients from 7 unrelated Italian families. In addition to clinical examination and routine laboratory testing, we measured bleeding time, platelet aggregation and release reaction, and platelet staining for tubulin, and performed ultrastructural study of polymorphonuclear leukocytes. RESULTS: Although the mean age of our patients was 33 years, May-Hegglin anomaly had not been previously recognized in any of them. Bleeding diatheses ranged from severe to absent, and platelet count from 26 to 178 x 10(9)/L. No correlation was found between bleeding tendency and platelet count. Previous therapy with corticosteroids, high-dose immunoglobulins, and splenectomy had no effect on platelet count or bleeding diathesis. Desmopressin infusion greatly shortened the bleeding time in the most severely affected patient. The in vitro function of platelets was normal except for the absence of shape change in all subjects and defective response to epinephrine in 8 of 15 patients. Platelet tubulin was distributed unevenly instead of being organized in a circumferential band at the cell periphery. CONCLUSION: The diagnosis of May-Hegglin is easily missed, and its frequency is probably underestimated. A qualitative defect of platelets may be responsible for mild bleeding diathesis even in the absence of thrombocytopenia, while severe bleeding results from both qualitative and quantitative platelet defects. May-Hegglin anomaly should be suspected whenever a patient has a low platelet count or a bleeding diathesis of unknown origin.


Subject(s)
Blood Platelets/pathology , Inclusion Bodies/pathology , Neutrophils/pathology , Thrombocytopenia/diagnosis , Adolescent , Adult , Aged , Bleeding Time , Child, Preschool , Diagnosis, Differential , Female , Fluorescent Antibody Technique, Indirect , Humans , Male , Middle Aged , Platelet Aggregation
13.
Thromb Haemost ; 73(4): 689-92, 1995 Apr.
Article in English | MEDLINE | ID: mdl-7495080

ABSTRACT

We found that intracellular Ca2+ concentration ([Ca2+]i) increased during ristocetin-induced agglutination of aequorin loaded platelets resuspended in plasma. Chelation of extracellular Ca2+ had no effect on platelet clumping, but delayed and greatly reduced Ca2+ increase, indicating that it derived for the most part from Ca2+ influx. Nine monoclonal antibodies (MA) against glycoprotein (GP) Ib largely prevented ristocetin-induced platelet clumping and [Ca2+]i increase, while three anti-GPIb MA with no effect on platelet clumping did not interfere with Ca2+ movement. In unstirred samples platelet agglutination was greatly reduced and [Ca2+]i increase was abolished, suggesting that close platelet-to-platelet contact, in addition to von Willebrand factor (vWF) binding to GPIb, is necessary for Ca2+ transient. Nine MA against GPIIb/IIIa, the gly-arg-gly-asp-ser (GRGDS) peptide and GPIIb/IIIa complex dissociation had no effect on platelet agglutination, but significantly reduced Ca2+ increase. Our results suggest that platelet clumping induced by vWF binding to GPIb is responsible for GPIIb-IIIa dependent Ca2+ influx.


Subject(s)
Blood Platelets/physiology , Calcium/metabolism , Platelet Aggregation/drug effects , Platelet Glycoprotein GPIIb-IIIa Complex/metabolism , Platelet Glycoprotein GPIb-IX Complex/metabolism , Ristocetin/pharmacology , Calcium Channels/metabolism , Humans
14.
Thromb Haemost ; 68(2): 208-13, 1992 Aug 03.
Article in English | MEDLINE | ID: mdl-1412168

ABSTRACT

A young patient developed chronic idiopathic thrombocytopenic purpura. Prednisone therapy normalized platelet number, but bleeding symptoms did not disappear. Platelet function was severely impaired, since platelet aggregation, ATP release and adhesion to collagen and subendothelial matrix were significantly reduced. Plasma and purified immunoglobulins of the patient reproduced the functional defects in normal platelets. Immunoblotting revealed that patient's plasma contained an antibody reacting with a component of platelets with the same electrophoretic mobility of glycoproteins IIIa of normal platelets. Moreover, patient's plasma inhibited the binding of an anti-GPIIb/IIIa monoclonal antibody to platelet surface. Additional immunosuppressive therapy with prednisone and azathioprine normalized platelet function and induced the disappearance of bleeding symptoms.


Subject(s)
Autoantibodies/blood , Blood Platelet Disorders/immunology , Platelet Membrane Glycoproteins/immunology , Adult , Azathioprine/therapeutic use , Blood Platelet Disorders/blood , Blood Platelet Disorders/drug therapy , Female , Humans , Platelet Adhesiveness , Platelet Aggregation , Prednisone/therapeutic use , Purpura, Thrombocytopenic, Idiopathic/blood , Purpura, Thrombocytopenic, Idiopathic/drug therapy , Purpura, Thrombocytopenic, Idiopathic/immunology
15.
Am J Cardiol ; 69(5): 457-61, 1992 Feb 15.
Article in English | MEDLINE | ID: mdl-1736606

ABSTRACT

Whereas in vitro studies showed that plasmin may induce both inhibition and activation of platelets, in vivo and ex vivo investigations suggested that thrombolytic agents are responsible for platelet stimulation. To gain further information on this topic, ex vivo platelet function was studied in 24 subjects with acute myocardial infarction treated with streptokinase or recombinant tissue-type plasminogen activator (rt-PA). Ten patients with acute myocardial infarction who did not receive thrombolytic treatment were also investigated. The data shows that at the end of thrombolytic infusion, the maximal extent of platelet aggregation and adenosine triphosphate release was reduced in treated patients compared with that in untreated ones. In subjects treated with streptokinase, the defect in platelet aggregation derived from both cellular and plasmatic defects. Plasmatic beta-thromboglobulin concentration was significantly reduced after streptokinase, but unchanged after rt-PA. Three days after thrombolytic treatment, platelet aggregation of patients receiving streptokinase or rt-PA was not significantly different from that of untreated subjects. A similar defect in platelet function was obtained in vitro, incubating normal platelet-rich plasma with pharmacologic concentrations of streptokinase. Again, platelet function defect derived from both cellular and plasmatic damages. It cannot be excluded that platelet activation occurs in patients with acute myocardial infarction during the very early phases of thrombolytic treatment. However, it is suggested that a transient defect in platelet function follows both streptokinase and rt-PA infusion.


Subject(s)
Blood Platelets/drug effects , Myocardial Infarction/blood , Myocardial Infarction/drug therapy , Streptokinase/therapeutic use , Thrombolytic Therapy , Tissue Plasminogen Activator/therapeutic use , Adenosine Triphosphate/blood , Blood Platelets/physiology , Humans , In Vitro Techniques , Platelet Aggregation/drug effects , Time Factors
16.
Am J Clin Pathol ; 95(1): 82-6, 1991 Jan.
Article in English | MEDLINE | ID: mdl-1987755

ABSTRACT

In vitro platelet aggregation in platelet-rich plasma (PRP) and in whole blood (WB) was assessed in 31 patients with idiopathic myelofibrosis, 32 with essential thrombocytosis, 23 with polycythemia vera, and 34 with chronic myelogenous leukemia. In PRP most subjects showed normal or reduced platelet aggregation, whereas in WB the majority of patients showed increased platelet function. Spontaneous platelet aggregation (SPA) was observed frequently in WB, whereas it was seldom observed in PRP. SPA in WB was inhibited by in vitro addition of aspirin and apyrase, and SPA was only partially dependent on high platelet count because it also occurred in samples with normal platelet content (at variance with 13 subjects with reactive thrombocytosis, in which SPA was observed only in samples with high platelet concentration). Platelets from patients with idiopathic myelofibrosis had the highest tendency to undergo SPA.


Subject(s)
Myeloproliferative Disorders/blood , Platelet Aggregation , Adenosine Diphosphate/pharmacology , Collagen/pharmacology , Epinephrine/pharmacology , Humans , Platelet Aggregation/drug effects , Platelet Aggregation Inhibitors/pharmacology , Platelet Count , Thrombocytosis/blood
17.
Ital J Biochem ; 31(6): 428-34, 1982.
Article in English | MEDLINE | ID: mdl-7184907

ABSTRACT

Platelet membranes were isolated by the glycerol lysis technique and incubated in Krebs-Ringer phosphate buffer, to determine their chemical modifications occurring during "in vitro" incubation. The main changes observed at the end of the incubation consist in a significant loss of protein components and in the nearly equimolar decrease of sialic acid and galactosamine. Aminoacid analysis indicate that mainly polar aminoacids are lost. SDS-polyacrylamide gel electrophoresis indicates that high molecular weight proteins disappear after incubation, while no relevant differences in the relative ratios between PAS-stained bands were evident. The reported modifications are quite similar to those described in the membranes of in vivo and in vitro aged red cells.


Subject(s)
Blood Platelets/metabolism , Amino Acids/blood , Cell Membrane/analysis , Electrophoresis, Polyacrylamide Gel , Hexosamines/blood , Humans , Sialic Acids/blood
18.
Ann Ital Med Int ; 4(2): 83-8, 1989.
Article in Italian | MEDLINE | ID: mdl-2702024

ABSTRACT

We report on the effects of splenectomy in 45 consecutive patients affected by idiopathic thrombocytopenia who did not obtain lasting remission with prednisone therapy. None of the patients had serious complications after surgery, and only one died a year later from an infectious disease. Two years after surgery, 89% of the patients were in remission; 79% had no therapy after splenectomy, while 10% achieved remission in response to additional medical treatment. A significantly better response to splenectomy was obtained in the subjects under 50 years old (90% remission) and by the younger patients who had had a transient response to initial prednisone therapy (100% remission). All patients with thrombocytosis 15 days after surgery were in remission two years later. We were unable to identify parameters constantly associated with a poor response to splenectomy, since at least 25% of the patients with the worst prognostic features obtained lasting remission.


Subject(s)
Purpura, Thrombocytopenic/surgery , Splenectomy , Adolescent , Adult , Age Factors , Aged , Evaluation Studies as Topic , Female , Humans , Male , Middle Aged , Prednisone/therapeutic use , Prognosis , Purpura, Thrombocytopenic/drug therapy
19.
J Thromb Haemost ; 11(6): 1006-19, 2013 Jun.
Article in English | MEDLINE | ID: mdl-23510089

ABSTRACT

The diagnosis of inherited thrombocytopenias is difficult, for many reasons. First, as they are all rare diseases, they are little known by clinicians, who therefore tend to suspect the most common forms. Second, making a definite diagnosis often requires complex laboratory techniques that are available in only a few centers. Finally, half of the patients have forms that have not yet been described. As a consequence, many patients with inherited thrombocytopenias are misdiagnosed with immune thrombocytopenia, and are at risk of receiving futile treatments. Misdiagnosis is particularly frequent in patients whose low platelet count is discovered in adult life, because, in these cases, even the inherited origin of thrombocytopenia may be missed. Making the correct diagnosis promptly is important, as we recently learned that some forms of inherited thrombocytopenia predispose to other illnesses, such as leukemia or kidney failure, and affected subjects therefore require close surveillance and, if necessary, prompt treatments. Moreover, medical treatment can increase platelet counts in specific disorders, and affected subjects can therefore receive drugs instead of platelet transfusions when selective surgery is required. In this review, we will discuss how to suspect, diagnose and manage inherited thrombocytopenias, with particular attention to the forms that frequently present in adults. Moreover, we describe four recently identified disorders that belong to this group of disorders that are often diagnosed in adults: MYH9-related disease, monoallelic Bernard-Soulier syndrome, ANKRD26-related thrombocytopenia, and familial platelet disorder with predisposition to acute leukemia.


Subject(s)
Hematologic Diseases/diagnosis , Hematologic Diseases/genetics , Thrombocytopenia/diagnosis , Thrombocytopenia/genetics , Adult , Bernard-Soulier Syndrome/diagnosis , Blood Platelet Disorders/diagnosis , Blood Platelets/cytology , Diagnosis, Differential , Genetic Predisposition to Disease , Humans , Intercellular Signaling Peptides and Proteins , Leukemia/blood , Leukemia/genetics , Leukemia, Myeloid, Acute/diagnosis , Middle Aged , Molecular Motor Proteins/genetics , Myosin Heavy Chains/genetics , Nuclear Proteins/genetics , Platelet Count
20.
Hamostaseologie ; 32(4): 259-70, 2012.
Article in English | MEDLINE | ID: mdl-22972471

ABSTRACT

The chapter of inherited thrombocytopenias has expanded greatly over the last decade and many "new" forms deriving from mutations in "new" genes have been identified. Nevertheless, nearly half of patients remain without a definite diagnosis because their illnesses have not yet been described. The diagnostic approach to these diseases can still take advantage of the algorithm proposed by the Italian Platelet Study Group in 2003, although an update is required to include the recently described disorders. So far, transfusions of platelet concentrates have represented the main tool for preventing or treating bleedings, while haematopoietic stem cell transplantation has been reserved for patients with very severe forms. However, recent disclosure that an oral thrombopoietin mimetic is effective in increasing platelet count in patients with MYH9-related thrombocytopenia opened new therapeutic perspectives. This review summarizes the general aspects of inherited thrombocytopenias and describes in more detail MYH9-related diseases (encompassing four thrombocytopenias previously recognized as separate diseases) and the recently described ANKRD26-related thrombocytopenia, which are among the most frequent forms of inherited thrombocytopenia.


Subject(s)
Genetic Predisposition to Disease/genetics , Peptides/therapeutic use , Platelet Transfusion , Thrombocytopenia/congenital , Thrombocytopenia/therapy , Anticoagulants/therapeutic use , Humans , Thrombocytopenia/diagnosis
SELECTION OF CITATIONS
SEARCH DETAIL