Somatic Fas Mutations Account for Nearly One Third of Autoimmune Lymphoproliferative Syndrome (ALPS) Cases with Previously Unknown Genetic Mutations.

Blood ◽  
2009 ◽  
Vol 114 (22) ◽  
pp. 710-710
Author(s):  
Kennichi Dowdell ◽  
Julie Niemela ◽  
Susan Price ◽  
Joie Davis ◽  
Katie Perkins ◽  
...  

Abstract Abstract 710 Background: Autoimmune lymphoproliferative syndrome (ALPS) is characterized by childhood onset of lymphadenopathy, hepatosplenomegaly, autoimmune cytopenias, elevated (>1%) double negative T (DNT; CD3+, TCRalpha-beta+, CD4−, CD8−, B220+) lymphocytes in peripheral blood and an increased risk of lymphoma; primarily due to impaired lymphocyte apoptosis. Most cases (65%; 173 individuals in our cohort), known as ALPS Type Ia, are associated with dominant heterozygous germline mutations in the gene TNFRSF6 encoding the protein for CD95 (Fas, Apo-1). Another 5% of patients have mutations in FasL (Type Ib), caspases (Type II) or NRAS (Type IV). However, approximately 30% of ALPS patients in our cohort have no mutation found upon genomic sequencing of DNA from peripheral blood. We designated these patients as ALPS Type III (16%) if they met all the criteria of ALPS, i.e., elevated DNT cell numbers, nonmalignant chronic lymphadenopathy/splenomegaly and defective lymphocyte apoptosis by in vitro assay; we called them ALPS phenotype (14%) if they had all the features as described above but did not demonstrate an in vitro apoptosis defect. Holzelova et. al. (NEJM 2004; 351:1409) previously identified somatic Fas mutations in DNT cell population in 6 patients with ALPS phenotype. However, there was no information as to the proportion of ALPS patients expressing a somatic Fas mutation or any differences in their clinical phenotype. Methods and Results: Over the last 5 years we sought to determine the proportion of ALPS Type III and ALPS phenotype patients with somatic Fas mutations in their DNT cell population and to further clinically characterize these patients. DNT cells were purified to >50% by magnetic bead separation and DNA was sequenced for Fas. We found somatic Fas mutations in the DNT cells of 11/31 (35.5%) patients; 5/15 with ALPS Type III (5 males; age range = 1-17 years; median = 12 years), and 6/16 with ALPS phenotype (3 males and 3 females; age range = 3 mo–48 years; median age = 1 year). All the mutations clustered to an approximately 150 base region of the intracellular domain of Fas (exons 7, 8 and 9), except for one mutation in exon 3. All mutations resulted in functional loss of normal Fas signaling based on the mutation type or having been previously observed in ALPS Typa Ia. The somatic ALPS patients showed a similar clinical phenotype to that of ALPS Type Ia with increased DNT cell numbers (Median 6%; Range 4-19%), and increased levels of biomarkers like serum vitamin B12, IL-10 and sFasL. All 4 patients with onset of symptoms in infancy have required long-term treatment of their refractory autoimmune cytopenias with mycophenolate mofetil for the last 2-8years, while one of them underwent splenectomy. Inability to demonstrate defective lymphocyte apoptosis by in vitro testing of unseparated lymphocytes in ALPS phenotype patients with somatic Fas mutations is likely due to the relatively small number of affected cells (<20%) in their peripheral blood. Additionally, we hypothesize that the late onset of clinical disease in ALPS Type III patients with somatic Fas mutations may reflect the time required to accumulate a threshold number of cells that are able to confer clinical manifestations. Conclusion: Thus, the majority of somatic ALPS mutations can be detected by sequencing exons 7-9 of isolated DNT cell DNA. Patients with somatic Fas mutations now comprise the second largest group of known genetic mutations in ALPS, followed by patients with caspase 10, NRAS, caspase 8 and FasL germline mutations, respectively. We recommend testing for somatic Fas mutations in ALPS Type III patients (particularly those with late onset) and all ALPS phenotype patients, using ungated DNT purities of >50%. These data also highlight the role of somatic mutations in the pathogenesis of nonmalignant hematological conditions in adults and children, as well as in clonal malignancies. Disclosures: No relevant conflicts of interest to declare.

Blood ◽  
2005 ◽  
Vol 106 (11) ◽  
pp. 2396-2396
Author(s):  
V. Koneti Rao ◽  
Kennichi C. Dowdell ◽  
Faith Dugan ◽  
Janet K. Dale ◽  
Lesley Pesnicak ◽  
...  

Abstract ALPS is an inherited disorder of lymphocyte apoptosis leading to childhood onset of chronic lymphadenopathy, hepatosplenomegaly, autoimmunity, and an increased risk of lymphoma in a subset of patients with mutations in the intracellular domain of Fas (ALPS Type Ia). Similarly, MRL/lpr −/− mice are homozygous for Fas mutations and develop massive lymphadenopathy and splenomegaly associated with hypergammaglobulinemia, glomerulonephritis, and expansion of TCR αβ+, CD4−/CD8−double-negative (DN) T cells that are pathognomonic of ALPS. There remain no proven therapies for the lymphoproliferation underlying ALPS itself, although investigators in Europe reported that some children with ALPS showed reductions in spleen and lymph node sizes while on a weekly regimen of pyrimethamine/sulfadoxine (Fansidar@) (Br J Haematol.2002; 117:176). However, in vitro studies using stimulated human PBMCs, revealed that lymphocyte apoptosis is induced by the antifolate agent, pyrimethamine (Pyr) at an EC50 of 2.5ug/mL, and not sulfadoxine. This suggested that Pyr alone might prove clinically beneficial, thus avoiding risks of allergy and hypersensitivity associated with sulfa drugs. Hence, studies were conducted in the MRL/lpr−/− murine model of ALPS with Pyr alone given 2mg/kg twice weekly by gavage for 8 weeks, and in combination with sulfadoxine. Neither Pyr nor Fansidar yielded significant shrinkage of lymphoid mass in mice. A pilot study of the safety and efficacy of Pyr was conducted under an FDA IND in 6 children with ALPS Type 1a and one with ALPS Type III (with no identifiable mutation). Aged 6 to 17 yrs (median; 13 y), these 6 males and 1 female patient were enrolled following informed consent to be treated with oral Pyr at escalating doses (25–50mg) given twice weekly for 12 weeks. Computerized tomography was done before and after treatment to determine whether these patients would show 40 and 50% reductions in the sizes of their lymphadenopathy and/or splenomegaly, respectively. Lymph node sizes were assessed as sums of the bi-dimensional products of 3 to 5 nodes. Spleen sizes were measured as products of their craniocaudal and anterioposterior dimensions at the superior mesenteric artery level, as well as spleen volumes computed by digital image processing. The effects of treatment on other well-defined laboratory features of ALPS were also assessed. Pyr was discontinued after 11 days in one patient due to skin rash. Dose escalation was interrupted transiently in 3 others because of neutropenia, aphthous ulcer, or leucopenia. There were no significant changes in study outcome measures. The aggregate experience with Pyr and Fansidar in mice, as well as with Pyr alone in ALPS patients (Table) lead us to conclude that these agents do not manifest sufficient effectiveness to warrant larger trials.


Blood ◽  
2006 ◽  
Vol 108 (11) ◽  
pp. 3900-3900 ◽  
Author(s):  
Kennichi C. Dowdell ◽  
Julie Niemela ◽  
Janet K. Dale ◽  
Jennifer Puck ◽  
V. Koneti Rao ◽  
...  

Abstract ALPS is a disorder of lymphocyte apoptosis associated with expansion of double negative T cells (DNT; TCRα/β+CD3+CD4−CD8−), an in vitro apoptosis defect and affected individuals typically present in early childhood with nonmalignant lymphadenopathy, splenomegaly, and multilineage cytopenias due to splenic sequestration and/or peripheral autoimmune destruction. ALPS classifications include Type Ia, Ib, and II associated with mutations in Fas, FasL, and Caspases, respectively, and Type III when the apoptosis pathway mutation is unidentified. Among our cohort of more than 200 patients, ALPS Type Ia with germline Fas mutations constitute the most common subtype (over 70%). These patients show an in vitro Fas mediated lymphocyte apoptosis defect and have a known increased risk of lymphoma. However, there are a number of patients that have an “ALPS-phenotype” manifesting typical historical and clinical features of ALPS and present with elevated circulating DNT cells, but have no demonstrable in vitro apoptosis defect. Based on the work of Holzelova et. al (NEJM, 2004, 351:1409) in which six such patients were reported with somatic Fas mutations in their DNT cell subsets, we sought to characterize our patient set without defined mutations. DNT cells from 20 patients, including 8 with ALPS-phenotype and 12 with ALPS Type III, were isolated using magnetic bead separation of frozen PBMCs. The sensitivity for PCR followed by sequencing required >20% abnormal cells using a mixture of normal and ALPS Type Ia PBMCs. However, the sensitivity proved to be lower (40%) using purified DNT cells from patients with potential somatic Fas mutations. Therefore, we chose an ungated DNT purity of 50% as a minimum requirement. Ungated DNT purities for the 20 evaluated patients ranged from 51% to 92%, with a mean of 66%. All nine exons of the Fas gene were PCR amplified prior to sequencing. We found that 3/8 (37.5%) patients with ALPS-phenotype had somatic mutations in the Fas gene: 825G−>T, 846(−9)del11, and 846(−1)G−>T, with the first two mutations being new. Only 1/12 (8.3%) ALPS Type III patients had a somatic Fas muation: 851delAG, which also represents a new mutation. These four somatic mutations were considered significant as two were in a splice site and two were in coding regions. The ALPS Type III patient is atypical given a late age of presentation at age 17 and had milder cytopenias. The three ALPS-phenotype patients with somatic Fas mutations presented similar to cases of “classic” ALPS, with an early age of presentation (median, 1 yr; range, 4 months to 4 years), cytopenias and features of autoimmunity; two require chronic immunosuppression with Cellcept (mycophenolate mofitil) for chronic persistent cytopenias, one having previously undergone surgical splenectomy. From these data we conclude that patients with ALPS-phenotype or atypical ALPS Type III should be screened for somatic mutations in their peripheral blood DNT cell subset using ungated DNT purities of >50%, with priority given to evaluation by sequencing of exons 7–9 of the Fas gene. It is not yet evident if there is an increased risk of lymphoma in patients with somatic Fas mutations as is the case with intracellular germline Fas mutations. Somatic Fas mutations explain the molecular basis of a small subset of patients and are more readily observed in ALPS-phenotype patients.


Blood ◽  
2007 ◽  
Vol 110 (11) ◽  
pp. 1385-1385 ◽  
Author(s):  
Kennichi C. Dowdell ◽  
Lesley Pesnicak ◽  
Lilia Bi ◽  
Victoria Hoffmann ◽  
V. Koneti Rao ◽  
...  

Abstract Hydroxychloroquine (HCQ) is an anti-malarial drug in clinical use for decades that is finding further use as a steroid sparing agent in the treatment of immune disorders such as chronic GVHD, lupus and rheumatoid arthritis. HCQ is a lysosomotropic agent with more recent evidence showing immunomodulatory anti-TNF activity. It is currently being explored as a cytotoxic antineoplastic/antimicrobial agent. Hence, studies were conducted to determine the efficacy of HCQ to control the lymphoproliferation associated with ALPS. ALPS is an inherited disorder of apoptosis leading to lymphoproliferation and autoimmunity. The majority of ALPS patients are classified as Type Ia (>70%), having germline mutations in Fas. Other ALPS patients are classified as Type Ib, II, IV or III, if they have mutations in FasL, casapases, NRAS, or no identified mutations, respectively. They often present with childhood onset autoimmune cytopenias with lymphadenopathy, splenomegaly, increased circulating double negative T cells (DNT; TCRa/b+CD3+CD4−CD8−), defective apoptosis in vitro, and have an increased risk of lymphoma. Similarly, MRL/lpr−/− mice homozygous for Fas mutations develop an ALPS-like disease with massive lymphadenopathy, splenomegaly, hypergammaglobulinemia, autoimmune glomerulonephritis, and expansion of DNT cells secondary to defective lymphocyte apoptosis leading to lymphomagenesis. Currently, there are no proven therapies for the lymphoproliferation underlying ALPS itself. PBMCs from normal controls and ALPS Type Ia patients were cultured in vitro with 0–120 ug/mL HCQ in the presence or absence of 50 uM of the pan-caspase inhibitor Z-VAD-FMK, the caspase 9 inhibitor Z-LEHD-FMK, or the caspase 8 inhibitor Z-IETD-FMK. A dose response was observed with a high degree of cell death noted at 120 ug/mL after 48 hours, with an LD50 of 40 ug/mL. HCQ induced cell death was through a caspase-independent mechanism based on no inhibition of cell death by Z-VAD-FMK, Z-LEHD-FMK, or Z-IETD-FMK. Further preclinical studies were conducted in the MRL/lpr−/− murine model of ALPS. Forty five, 8-week old female MRL/lpr−/− mice were treated with 30 or 60 mg/kg of HCQ by gavage in sterile PBS or PBS alone three days per week for up to 14 weeks. Reductions of the spleen weight (p = 0.045, 437±85 vs 240±27) and cellularity (p = 0.08, 217±32 vs 148±18) were observed in high dose HCQ treated mice compared to controls at 7 weeks. Reductions in cellularity were also noted in the lymph nodes (p = 0.032, 118±33 vs 38±6). There was a trend towards decreasing DNT percentages in the spleen (21±5 vs 14±2), LN (70±4 vs 67±2) and blood (57±6 vs 51±5). Additionally, the CBC showed only marginal reductions in the WBC count (4.2±0.5 vs 3.5±0.3), hematocrit (49±0.3 vs 46±1.0), polymorphonuclear cells (33±3.7 vs 29±3.5) and monocytes (23±3.2 vs 17±2.5). Future studies are planned to more clearly determine the effect of HCQ on autoimmune kidney disease. Based on our in vitro and in vivo data, HCQ is effective at reducing lymphoproliferative activity in Fas deficient MRL/lpr−/− mice. It is being further explored in early phase clinical trials as a lympholytic agent to shrink lymph nodes and abrogate hypersplenism in ALPS patients. HCQ may also prove beneficial for long term use, especially in children, as a steroid sparing agent for treating refractory autoimmune cytopenias in ALPS.


2020 ◽  
Vol 14 (Supplement_1) ◽  
pp. S149-S150
Author(s):  
H Kiyohara ◽  
T Toyonaga ◽  
S Kuronuma ◽  
A Ueno ◽  
S Okabayashi ◽  
...  

Abstract Background A novel thiopurine metabolizing enzyme, nucleotide diphosphate-linked moiety X-type motif 15 (NUDT15) was associated with drug-induced leukopenia in patients with non-synonymous genetic polymorphisms. Thiopurine-induced leukopenia in Japanese patients with genetic variance in NUDT15 (c.415C&gt;T) appears to be independent of the 6-thioguanine nucleotide concentration in red blood cells. However, detailed molecular mechanism how NUDT15 variance causes thiopurine-induced leukopenia remains unclear and NUDT15-associated subcellular thiopurine metabolism has not been investigated in patients with inflammatory bowel diseases (IBD). Methods DNA-incorporated deoxythioguanosine (dTG) was measured in the peripheral blood mononuclear cells (PBMCs) of Japanese patients with IBD under thiopurine treatment. Association of a single-nucleotide polymorphism for NUDT15 (c.415C&gt;T) with dTG in PBMCs (dTGPBMC) was examined. Peripheral blood T lymphocytes were cultured in vitro with 6-thioguanine (6-TG) to examine the Impact of NUDT15 genotypes on incorporation into DNA, cell proliferation and apoptosis. Results NUDT15 variants had significantly higher dTGPBMC per thiopurine dosage than non-variants (homozygous variants (TT) vs. heterozygous variants (CT) vs. non-variants (CC), 4418.0 vs. 663.0 vs. 295.3 dTG mol/106 moles dA per mg/kg/day of 6-MP (Figure A)). dTGPBMC and peripheral lymphocyte counts showed a negative correlation (r = −0.30, p = 0.015) (Figure B). Peripheral blood lymphocytes from patients with NUDT15 variance showed a higher DNA-incorporated dTG associated with increased apoptosis (increase of Annexin V+ PI+ CD4+ lymphocytes; TT vs. CT vs. CC, 158.5 % vs. 80.1 % vs. 57.9 % (p = 0.0427)) (Figure C) and decreased proliferation (decrease of proliferative CD4+ lymphocytes, TT vs. CT vs. CC, 49.0 % vs. 25.0 % vs. 19.1 % (p = 0.0098)) (Figure D) when cultured with 6-thioguanine in vitro. Conclusion DNA-incorporated dTG affected by NUDT15 genotypes induces T lymphocyte apoptosis in patients with IBD.


Blood ◽  
2006 ◽  
Vol 108 (11) ◽  
pp. 2497-2497
Author(s):  
Kennichi C. Dowdell ◽  
Lesley Pesnicak ◽  
Victoria Hoffman ◽  
Kenneth Steadman ◽  
Mark Ruddel ◽  
...  

Abstract ALPS is an inherited disorder of apoptosis leading to lymphoproliferation and autoimmunity. ALPS Type Ia, Ib and II are associated with germline mutations in Fas, FasL and Casapase 8 or 10, respectively; patients in whom no mutations have been identified are classified as Type III. The vast majority of patients are ALPS Type Ia (greater than 70%). They often present with childhood onset autoimmune cytopenias associated with lymphadenopathy, splenomegaly, increased double negative T cells (DNT; TCRα/β+CD3+CD4−CD8−), defective apoptosis by in vitro assay, and have an increased risk of lymphoma. Similarly, MRL/lpr−/− mice homozygous for Fas mutations develop an ALPS-like disease with massive lymphadenopathy, splenomegaly, hypergammaglobulinemia, autoimmune glomerulonephritis, and expansion of DNT cells secondary to defective lymphocyte apoptosis leading to lymphomagenesis. Currently, there are no proven therapies for the lymphoproliferation underlying ALPS itself, although complications like autoimmune cytopenias and post-splenectomy sepsis are manageable. Hence, studies were conducted to determine the efficacy of valproic acid (VPA) to control the lymphoproliferation associated with ALPS. VPA is a histone deacetylase (HDAC) inhibitor in clinical use for the last four decades as an anticonvulsant in children and adults, and recently being explored as an anti-neoplastic agent. PBMCs from normal controls and ALPS Type Ia patients were cultured in vitro with 0–4 mM VPA in the presence or absence of 50 uM of the pan-caspase inhibitor Z-VAD-FMK. A dose response was observed with a high degree of cell death noted at 4 mM after 48 hours, with an LD50 of 2 mM. VPA appeared to induce cell death by both caspase-dependent and -independent mechanisms based on partial inhibition of VPA-induced cell death by Z-VAD-FMK. Further preclinical studies were conducted in the MRL/lpr−/− murine model of ALPS. Twenty, 8-week old female MRL/lpr−/− mice were treated intraperitoneally (i.p.) with 500 mg/kg of VPA in sterile PBS or PBS alone five days per week for 8 weeks. Significant reduction of the spleen weight (p=0.034) and cellularity (p=0.0001) was observed in VPA treated (n=10) mice compared to controls (n=10). Reductions in size and cellularity were also observed in the lymph nodes (p=0.09 and 0.0002, respectively). A concomitant decrease (p<0.05) in DNT cells was observed in the spleen (11.2±0.6 vs 8.1±0.4) and blood (9.3±0.96 vs 5.5±0.6). Serum drug levels peaked (462±10 ug/mL) by 2 hours post-i.p. injection of VPA, where-as a 2.5 fold increase in histone acetylation was observed in the spleen at 4 hours, following a single injection. Analysis of the effects of VPA on autoimmune renal disease in these animals is underway. Based on our in vitro and in vivo data, VPA is effective at reducing lymphoproliferative activity in Fas deficient MRL/lpr−/− mice. It is being further explored in early phase clinical trials as a lympholytic agent to shrink lymph nodes and abrogate hypersplenism in ALPS patients.


1998 ◽  
Vol 72 (11) ◽  
pp. 9025-9033 ◽  
Author(s):  
Edgar Holznagel ◽  
Regina Hofmann-Lehmann ◽  
Christian M. Leutenegger ◽  
Karin Allenspach ◽  
Silke Huettner ◽  
...  

ABSTRACT Human immunodeficiency virus infection is characterized by a progressive decline in the number of peripheral blood CD4+T lymphocytes, which finally leads to AIDS. This T-cell decline correlates with the degree of in vitro-induced lymphocyte apoptosis. However, such a correlation has not yet been described in feline AIDS, caused by feline immunodeficiency virus (FIV) infection. We therefore investigated the intensity of in vitro-induced apoptosis in peripheral blood lymphocytes from cats experimentally infected with a Swiss isolate of FIV for 1 year and for 6 years and from a number of long-term FIV-infected cats which were coinfected with feline leukemia virus. Purified peripheral blood lymphocytes were either cultured overnight under nonstimulating conditions or stimulated with phytohemagglutinin and interleukin-2 for 60 h. Under stimulating conditions, the isolates from the infected cats showed significantly higher relative counts of apoptotic cells than did those from noninfected controls (1-year-infected cats, P = 0.01; 6-year-infected cats, P = 0.006). The frequency of in vitro-induced apoptosis was inversely correlated with the CD4+ cell count (P = 0.002), bright CD8+ cell count (P = 0.009), and CD4/CD8 ratio (P = 0.01) and directly correlated with the percentage of bright major histocompatibility complex class II-positive peripheral blood lymphocytes (P = 0.004). However, we found no correlation between in vitro-induced apoptosis and the viral load in serum samples. Coinfection with feline leukemia virus enhanced the degree of in vitro-induced apoptosis compared with that in FIV monoinfected cats. We concluded that the degree of in vitro-induced apoptosis was closely related to FIV-mediated T-cell depletion and lymphocyte activation and could be used as an additional marker for disease progression in FIV infection.


Blood ◽  
1997 ◽  
Vol 89 (4) ◽  
pp. 1341-1348 ◽  
Author(s):  
Michael C. Sneller ◽  
Jin Wang ◽  
Janet K. Dale ◽  
Warren Strober ◽  
Lindsay A. Middelton ◽  
...  

Abstract Programmed cell death (apoptosis) of activated lymphocytes is critical to immune homeostasis. The cell surface protein Fas (CD95) and its ligand play a pivotal role in regulating lymphocyte apoptosis, and defective expression of either Fas or Fas ligand results in marked over accumulation of mature lymphocytes and autoimmune disease in mice. The results of recent studies suggest that defective lymphocyte apoptosis caused by mutations of the Fas gene can result in a severe autoimmune lymphoproliferative syndrome (ALPS) in humans. To define the clinical, genetic, and immunologic spectrum of ALPS, 9 patients and their families were extensively evaluated with routine clinical studies, lymphocyte phenotyping, genotyping, and in vitro assays for lymphocyte apoptosis. Individual patients were followed up for 3 months to 6 years. ALPS was identified in 9 unrelated children as manifested by moderate to massive splenomegaly and lymphadenopathy, hypergammaglobulinemia, autoimmunity, B-cell lymphocytosis, and the expansion of an unusual population of CD4−CD8− T cells that express the α/β T-cell receptor (TCR). All patients showed defective lymphocyte apoptosis in vitro. Heterozygous mutations of the Fas gene were detected in 8 patients. One ALPS patient lacked a Fas gene mutation. Healthy relatives with Fas mutations were identified in 7 of 8 ALPS kindreds. These relatives also showed in vitro abnormalities of Fas-mediated lymphocyte apoptosis, but clinical features of ALPS were not present in the vast majority of these individuals. ALPS is a unique clinical syndrome in which in vitro abnormalities of lymphocyte apoptosis are associated with abnormal lymphoproliferation and autoimmunity. These findings provide evidence that apoptosis of activated lymphocytes is an important mechanism for maintaining immunologic homeostasis and self-tolerance in humans. Fas gene mutations account for impaired lymphocyte apoptosis in only a subset of patients with ALPS.


2010 ◽  
Vol 207 (2) ◽  
pp. 177-183 ◽  
Author(s):  
Pierre Roubert ◽  
Béatrice Dubern ◽  
Pascale Plas ◽  
Cécile Lubrano-Berthelier ◽  
Rohia Alihi ◽  
...  

Human melanocortin 4 receptor (hMC4R) mutations with in vitro functional effects are responsible for 0.5–2.5% of severe obesity. Designing ligands that are able to counteract this in vitro-associated molecular defect is crucial to develop specific anti-obesity drugs in these genetically associated cases. We analyzed the in vitro effect of two novel melanocortin agonists, IRC-022493 and IRC-022511, on typical hMC4R mutations chosen based on the nature of their functional alterations, i.e. intracytoplasmic retention and/or reduced basal activity and/or reduced α-MSH potency. We assessed the in vitro ability of IRC-022493 and IRC-022511 to bind and activate hMC4R mutants. These mutations were found earlier in 11 obese French patients (median age (range) was 17.6 years (5.7–48.0) and body mass index (BMI)-Z-score 4.2 s.d. (1.5–5.5). The MC4R agonists were responsible for a significant activation of mutated hMC4R depending on the functional characteristics of the mutations. Both agonists were able to activate mutated hMC4R with decreased α-MSH potency, associated with or without decreased basal activity, to the same extent than α-MSH in wild-type MC4R. This result suggests that those mutations would be the best targets for the MC4R agonists among MC4R mutation-bearing obese patients. No specific clinical phenotype was associated with the differential response to pharmacological agonists. We identified two novel melanocortin agonists that were able in vitro to efficiently activate mutated hMC4R with impaired endogenous agonist functional response. These results stimulate interest in the development of these drugs for hMC4R mutations-associated obesity.


Blood ◽  
2010 ◽  
Vol 115 (25) ◽  
pp. 5164-5169 ◽  
Author(s):  
Kennichi C. Dowdell ◽  
Julie E. Niemela ◽  
Susan Price ◽  
Joie Davis ◽  
Ronald L. Hornung ◽  
...  

Abstract Autoimmune lymphoproliferative syndrome (ALPS) is characterized by childhood onset of lymphadenopathy, hepatosplenomegaly, autoimmune cytopenias, elevated numbers of double-negative T (DNT) cells, and increased risk of lymphoma. Most cases of ALPS are associated with germline mutations of the FAS gene (type Ia), whereas some cases have been noted to have a somatic mutation of FAS primarily in their DNT cells. We sought to determine the proportion of patients with somatic FAS mutations among a group of our ALPS patients with no detectable germline mutation and to further characterize them. We found more than one-third (12 of 31) of the patients tested had somatic FAS mutations, primarily involving the intracellular domain of FAS resulting in loss of normal FAS signaling. Similar to ALPS type Ia patients, the somatic ALPS patients had increased DNT cell numbers and elevated levels of serum vitamin B12, interleukin-10, and sFAS-L. These data support testing for somatic FAS mutations in DNT cells from ALPS patients with no detectable germline mutation and a similar clinical and laboratory phenotype to that of ALPS type Ia. These findings also highlight the potential role for somatic mutations in the pathogenesis of nonmalignant and/or autoimmune hematologic conditions in adults and children.


Sign in / Sign up

Export Citation Format

Share Document