Briefly, after partial tracheal resection under deep anaesthesia,

Briefly, after partial tracheal resection under deep anaesthesia, a 22-gauge catheter was inserted into the choana towards the heads of a portion of mice. Each nasal cavity was gently irrigated by l ml of sterile saline. Nasal lavage fluid (NLF) was collected and centrifuged, and the supernatant was stored at −20 °C for cytokines analysis using enzyme-linked immunosorbent assay (ELISA). Cytokine levels of IL-5, selleck inhibitor IL-10,

IL-17, TGF-β1, IFN-γ and endogenous LF in NLF were measured by ELISA according to the manufacturers’ instructions (Boster Biotech, Wuhan, China). The detection sensitivity of the ELISA kits was <2 pg/ml for all cytokines. Five mice per group were chosen for histopathology. Animals were decapitated

and the heads were decalcified, embedded in paraffin and sectioned as previously described [22]. Histological changes in the nasal mucosa of all groups were examined using haematoxylin-eosin (HE) staining for eosinophils and periodic acid-schiff stain (PAS) for goblet cells. The cytoplasm of eosinophils in the nasal lamina propria (LP) stains red by HE, while the cytoplasm of goblet cells from the epithelium stains purple by PAS. Eosinophils in the LP were counted in four different fields, and eosinophil frequencies were expressed as cells/mm2. Goblet cells were expressed as cells/mm of epithelium. Th1, Th2, Th17 and Treg cell transcription factor and cytokine mRNA expression levels were determined each group (n = 5 per group). Nasal mucosa from samples was obtained using toothed microscopic tweezers under a stereo microscope (ZAS301; Beijing, China) and immediately frozen at −70 °C. Total buy RG7420 RNA was extracted by Trizol (Invitrogen, Carlsbad, CA, USA), and 0.5 μg total RNA was used for the reverse transcription reaction using

a Rever Tra Ace qPCR RT Kit (TOYOBO, Osaka, Japan) according to the manufacturer’s instructions and as previously described [4]. The qPCR of T-bet (NM_019507.2), GATA-3 (NM_008091.3), ROR-C (NM_011281.2), FOXP3 (NM_001199348.1), IFN-γ (NM_008337.3), IL-5 (NM_010558.1), IL-17 (NM_010552.3), IL-10 (NM_010548.2), TGF-β1 (NM_011577.1), TNF-α (NM_013693.2) and LF (NM_008522.3) was performed with an ABI 7500 real-time PCR system (Applied Biosystems, Farnesyltransferase Foster City, CA, USA) using the SYBR qPCR mix (TOYOBO) according to the manufacturer’s protocol. Briefly, 1.0 μl cDNA was added to 10 μl SYBR qPCR mix, 7 μl RNase-free water and 1 μl of each primer (10 μm). The PCR conditions consisted of an initial denaturation at 95 °C for 50 s, followed by amplification for 40 cycles of 15 s at 95 °C, 15 s at 56–60 °C (varying between primer sets) and 50 s at 72 °C. An analysis of relative gene expression was calculated using the 2−ΔΔCT method on the ABI 7500 Sequence Detection System Software (Applied Biosystems). Gene expression was normalized to glyceraldehydes-3-phosphate dehydrogenase (GAPDH, NM_008084.2).

The implantation biopsy showed minimal transmitted mesangial IgA1

The implantation biopsy showed minimal transmitted mesangial IgA1

deposition. Immunosuppressive treatment was administered with basiliximab induction, tacrolimus, mycophenolate mofetil and steroids. At discharge, graft function was satisfactory (serum creatinine (sCr), 1.28 mg/dL), and the 24 h proteinuria was 0.32 g. The initial protocol biopsy performed 2 weeks after transplantation showed mesangial IgA2, but not IgA1, deposition by immunofluorescence (IF) staining. Based on the results of the native kidney biopsy performed at an outside institution, the patient X-396 price was diagnosed with probable recurrent IgAN. This finding persisted for 6 months after transplantation and a tonsillectomy was subsequently performed. One year post transplantation, sCr levels increased to 2.2 mg/dL with the appearance of Nivolumab subnephrotic proteinuria (2.03 g/day) and microhematuria. The third biopsy performed 1 year after transplantation revealed minimal mesangial and endocapillary proliferative glomerulonephritis, although there was no evidence of rejection. Twenty-one months after transplantation, the patient received a low-dose rituximab infusion (200 mg) without complications. Over the next 8 months, however, graft function gradually deteriorated, and could not

be controlled by rituximab. A further allograft biopsy performed at 2 years after transplantation showed moderate tubular atrophy and interstitial fibrosis with signs of glomerular mesangial expansion and focal segmental proliferative lesions in the glomeruli (Fig. 1A). The following additional laboratory data were obtained: IgA, 162 mg/dL; IgG, 627 mg/dL; IgM, 43 mg/dL. Test results for both hepatitis B and C and serum cryoglobulins were negative. Serum immunoelectrophoresis showed the presence

of IgA monoclonal paraproteins. A retrospective study of all allograft biopsies showed diffuse mesangial staining for IgA (IgA2 only), C3 and λ light-chain, with negative staining for κ light-chain on IF (Fig. 1B–F). Electron microscopy (EM) performed on the fourth biopsy revealed large, finely granular, electron-dense deposits without a defined structure that were located Cediranib (AZD2171) primarily in the paramesangial regions (Fig. 1G). The patient eventually returned to haemodialysis 31 months after transplantation. IgAN is the most common primary glomerular disease, and therefore, it is a common indication for kidney transplantation.[1] The diagnostic hallmark of IgAN is the predominance of IgA deposits in the glomerular mesangium on IF; the IgA deposits, which are usually polyclonal, were suggested to be predominantly of the λ type, and are rarely found in a monoclonal form.[2] The disease has diverse clinical manifestations, reflecting a wide range of histological changes.

In addition, Th17 cells can be converted into Th1 cells in differ

In addition, Th17 cells can be converted into Th1 cells in different animal

models 21, 22. Furthermore, human CD4+ Tregs can be converted to a Th2 cell lineage subsequent to decreased FOXP3 expression 23. More recent studies have shown that CD4+ Tregs can also differentiate into IL-17-producing Th17 cells (IL-17+FOXP3+), and Th17 cells can co-express FOXP3 and RORγt (RoRγt+FOXP3+) 24, 25. Although these studies have focused on Th17 and Treg commitment and plasticity, whether Th17 cells can reciprocally convert into Tregs has not been described. In addition, the majority of studies demonstrating the plasticity of T-cell development have been based on observations in mixed cell populations without clear proof that this occurs selleck inhibitor at the single-cell level. Further precise investigations of

plasticity and the intimate links between T-cell lineages at a homogeneous cell clonal level will be critical for better understanding of T-cell-mediated immunity. To further explore the phenotypic and functional features of human Th17 cells, we have recently generated Th17 clones from tumor-infiltrating T lymphocytes (TILs) which were characterized by their transcriptional factor expression, cytokine and chemokine receptor expression profiles, and their effector function. During the course of procedures intended to maintain the stability of Th17 clones for future studies, we

unexpectedly found that these Th17 clones could differentiate into IFN-γ-producing and MG-132 cost FOXP3+ cells after in vitro stimulation with OKT3 and allogeneic peripheral blood mononuclear cells (PBMCs). Further studies showed that this Th17-to-Treg differentiation was specifically due to T-cell receptor (TCR) stimulation and was associated with FOXP3 demethylation and reprogramming of gene expression signatures, including lineage-specific transcriptional factor and cytokine genes, in Th17 cells following TCR stimulation and expansion. In addition to the expression of IFN-γ and FOXP3, these Th17 clones exhibited potent suppressive function following three rounds of repetitive stimulations and expansions with OKT3 and allogeneic PBMCs, suggesting their differentiation into Tregs. We also demonstrated that these Th17-derived Tregs science were resistant to Th17 reconversion in the presence of Th17 differentiation cytokines, including IL-2, IL-1β, IL-6 and IL-23. These results further indicate the substantial developmental plasticity of human Th17 cells and provide the first evidence that human Th17 cells can differentiate into Tregs at a T-cell clonal level. In the course of studies to examine the role of TIL subsets in anti-tumor immunity, we observed increased numbers of CD4+ Th17 cell populations in tumors of melanoma, ovarian, breast and colon cancers 26, 27.

Our study suggests that Bcl-3 may be an effective target for prom

Our study suggests that Bcl-3 may be an effective target for promoting regeneration of the epithelium in the colon. Bcl3−/−C57BL/6 (B6) mice were generated as described previously [15, 16]. All mice were group-housed in individually ventilated cages (IVCs) under specific pathogen-free conditions. Standard Dorsomorphin clinical trial housing and environmental conditions were maintained (temperature

21°C, 12 h light/12 h darkness with 50% humidity). Animals were fed sterile standard pellet diet and water ad libitum. Animal husbandry and experimental procedures were approved by the University College Cork Animal Experimentation Ethics Committee (AEEC). Mice were administered 2% DSS (45 kDa; TdB Consultancy, Uppsala, Sweden) ad libitum in their drinking water to induce colitis, as described previously [18]. DSS solutions were prepared freshly

and administered on a daily basis for 6 days. This was followed by water up to day 8 to induce acute disease. Body weight, stool consistency and posture/fur texture were recorded daily to determine the daily disease activity index (DAI). DAI scoring was assessed blinded with a maximum score of 10, as described previously [18, 19]. DAI scoring combined scoring from weight loss (% change) 0–4, stool consistency 0–4 and posture/fur texture 0–2. Briefly, a percentage weight loss score of 0 = no loss, 1 = 1–3% loss, 2 = 3–6% loss, 3 = 6–9% loss and 4 = greater than 9% loss in body mass. A stool

consistency score of 0 = no change, 1 = mild change, 2 = loose stool, 3 = loose stool and rectal bleeding, 4 = diarrhoea and rectal EX 527 mw bleeding. A fur and posture score 0 = no change, 1 = mild hunched posture, 2 = hunched posture and reduced movement. Mice were killed at day 8 with colons removed from anus to caecum and washed in phosphate-buffered saline (PBS). Colons were measured and cut longitudinally dividing into the distal and proximal colon. Both proximal and distal colons were weighed and processed for histology, protein and quantitative reverse transcription–polymerase chain reaction (qRT–PCR) Paclitaxel mouse analysis. Distal colons (3 cm) were cut longitudinally and into three sections. One section was rolled in a ‘swiss roll’ fashion and frozen in optimal cutting temperature (OCT) tissue-freezing medium (Tissue Tek, Sakura Finetek, Torrance, CA, USA) using liquid nitrogen. Frozen sections (6 μm) were fixed in ice-cold acetone/ethanol 3:1 solution and stained with haematoxylin and eosin (H&E) according to standard histological staining procedures. Stained sections were analysed and scored using a light microscope (Olympus BX51; Olympus, Hamburg, Germany). Images were captured using Cell F software (Olympus). Images captured are representative of greater than seven fields of view at ×20 magnification per mouse. Histological scoring was performed in a blinded fashion.

Subjects with following cardiovascular diseases are also excluded

Subjects with following cardiovascular diseases are also excluded: stroke, AMI, coronary artery disease (CAD), eye thrombus, angina pectoris, frequent arrhythmia, AOD, phlebitis, or rheumatic fever. The donors were between 18 and 65 years old, and their haemoglobin levels 135–195 and 125–175 g/l for men and women, respectively. All subjects gave their informed consent. The Local Ethics Committee at Helsinki University Central Hospital and the Finnish Red Cross, Oulu, Finland approved the study protocol. Sera were separated, divided into aliquots, and stored at −20 °C. Serum matrix metalloproteinase-8 (MMP-8), tissue inhibitor

of MMP-1 (TIMP-1), MPO, and HNE concentrations were determined both in the patients with arterial disease

and MG-132 molecular weight in the serum of the reference subjects. MMP-1 and MMP-13 concentrations were determined only in the patients. MMP-1, MMP-8, MMP-13, and TIMP-1 concentrations were determined using commercially available enzyme-linked immuno-sorbent assay (ELISA) kit according to the manufacturer’s instruction (Biotrak ELISA System; Amersham Biosciences, Buckinghamshire, UK) [15]. MPO (Immundiagnostik AG, Bensheim, Germany) and HNE (Alexis Biochemicals, Bender MedSystems, Vienna, Austria) concentrations were also analysed by ELISA. The absorbances were measured at 450 nm using Labsystems Multiskan RC (Thermo Bioanalysis Corporation, Santa Fe, USA), and the concentrations RAD001 were expressed as ng/ml. Serum concentrations of high-sensitive C-reactive protein (hsCRP), high- (HDL) and low-density (LDL) lipoprotein cholesterol, triglycerides, total cholesterol, Chlamydia pneumoniae markers (C. pneumonia IgA, IgG, and lipopolysaccharide),

antibody levels to Aggregatibacter actinomycetemcomitans (IgA, IgG), Porphyromonas gingivalis (IgA, IgG), and human heat-shock protein 60 (HSP60, IgA, and IgG), total lipopolysaccharide (LPS), LPS-binding protein (LBP), interleukin-6 (IL-6), and CD14 in the patients were measured as described previously [16]. Molar ratio of MMP-8 and TIMP-1 (indicated as MMP-8/TIMP-1) was determined by dividing the concentrations with the corresponding molecular weights, 65,000 Da click here and 28,000 Da, respectively [17]. The statistical significance of the differences between the groups was analysed by the student’s t-test. Correlation analyses of serum MMP and regulator levels were performed separately with in the patients and the healthy subjects by scatter plots and Pearson correlation analysis. Owing to the heterogeneous nature of the study population, the comparisons were done between the subgroups as well. Continuous variables are presented as median (interquartile range, IQR of 25–75%).

This finding is consistent with the speculation [57] that intrave

This finding is consistent with the speculation [57] that intravenously administered DCs can acquire islet antigens in vivo (a process that would take place in the pancreatic lymph nodes) and, thus, can modulate effector and regulatory T cell responses to diabetes-relevant antigens even without deliberate prior antigen treatment. The original observation that DCs from the pancreatic

lymph nodes could prevent diabetes when transferred to NOD mice, while those from other sites could not, suggested the potential importance of the incorporation of beta cell antigens into DC-based therapeutics for this disease [5]. As reviewed recently [66], a variety of immunosuppressive and anti-inflammatory compounds, e.g. vitamin D3 and mycophenolate mofetil, PI3K Inhibitor Library in vivo can endow DCs with a tolerogenic functional phenotype. Cytokines such as IL-10 can behave similarly [67]

. This suggests a therapeutic strategy for type 1 diabetes in which tolerogenic DCs would be generated in GPCR Compound Library cell assay vitro and then exposed to beta cell antigens prior to administration. Such an approach was employed recently by the von Herrath group [59], who utilized the rat insulin promoter (RIP)-LCMV model of type 1 diabetes in which disease is induced upon LCMV infection. BMDCs were generated in the presence of GM-CSF, IL-10 and normal mouse serum, and then pulsed with a viral peptide recognized by CD8+ T cells. When the pulsed DCs were administered intraperitoneally to mice 10 and 3 days prior to LCMV infection, only 45% of the animals developed diabetes, whereas 80% of those treated with unpulsed DCs became diabetic. A reduced expansion of viral-specific T cells in response to viral infection was also observed

in mice treated with peptide-pulsed DCs. This study supports the idea that ex vivo-generated tolerogenic DCs, when exposed to disease-relevant antigens, can deliver therapeutic benefit in type 1 diabetes. In a recent thoughtful review of DC-based immunotherapeutic strategies for human diseases, the disadvantages of ex vivo antigen loading of DCs were discussed [68]. These include a requirement for leukapheresis, the inability to manipulate DCs N-acetylglucosamine-1-phosphate transferase within their natural milieu and a requirement for a tailor-made ‘product’ for each patient, resulting in labour-intensive procedures and high costs. It is for reasons such as these that we [69] and others [70] are exploring the utility of in vivo delivery of beta cell antigens to DCs in the prevention and treatment of type 1 diabetes. DCs employ a variety of molecules, such as the Fc receptors, the macrophage mannose receptor (MMR) and DEC-205 [71], to execute receptor-mediated endocytosis of antigens. Of these, DEC-205 (Ly75/CD205) has the special ability to uptake and subsequently present antigen via both class I [35] and class II MHC pathways [72]. DEC-205 is a type 1 transmembrane protein homologous to MMR and phospholipase A2 [71].

Lactoferrin, a component of

breast milk and genital secre

Lactoferrin, a component of

breast milk and genital secretions, has also been shown to inhibit HIV-1 CYC202 cell line replication and transmission from dendritic cells (DCs) to T cells in vitro[70–72]. Nevertheless, cervicovaginal levels of lactoferrin, RANTES and SLP1 were tested in HIV-1 seronegative women at a high risk of heterosexual acquisition of HIV infection and were found to be associated with bacterial vaginosis and inflammation rather than exposure to HIV-1 [73]. In contrast, elafin/trappin-2 was found to be elevated in the female genital tract of HESN Kenyan sex workers and was associated with protection against HIV-1 acquisition [74]. Defensins represent a family of small cationic peptides expressed in the mucosal epithelium with broad anti-microbial properties against HIV-1 and other sexually transmitted diseases relevant to HIV-1 transmission [75]. Both α-defensins and Dorsomorphin datasheet β-defensins have been associated repeatedly with

protection in several independent studies of HESN subjects [76–80]. This includes the description of alpha-defensins in the prevention of HIV transmission among breastfed infants [76] and the identification of elevated levels of both alpha and beta-defensins in sexually HIV-1 exposed but uninfected individuals [79,80]. Despite potent HIV inhibitory activity, however, cervicovaginal levels of α-defensins have also been associated with increased HIV acquisition due to their association with bacterial sexually transmitted infections [81]. The role of α-defensins in HIV-1 vertical transmission remains contentious, with one study showing no association between α-defensin concentration in breast milk and risk of HIV-1 transmission [82] while another study showed the opposite [76]. Overall, the varied secreted proteins identified in the mucosa of HESN subjects (summarized in Table 2) may represent true factors associated with reducing

mucosal transmission of HIV-1 infection. Rather, they may reflect the innate immune response to genital tract inflammation due to ongoing bacterial infections or sexually transmitted diseases, which may be endemic in the case of sex worker G protein-coupled receptor kinase cohorts. Taking the data as a whole, we interpret that soluble innate factors are likely to modulate the infectivity threshold for HIV-1 upon exposure. However, secreted anti-viral factors alone are unlikely to render a complete barrier to infection, and innate immune cells such as natural killer (NK) cells and DCs may also bolster the threshold to infection that HIV-1 must overcome. NK cells represent a critical component of the host innate immune response against viral infection and serve as a front-line defence against a diverse array of pathogens.

Thus, we do not exclude that, in SN-APS

Thus, we do not exclude that, in SN-APS selleck chemical patients, phospholipid-binding proteins may also be involved in anti-phospholipid reactivity, as TLC immunostaining does not exclude this possibility. However, at present the involvement of phospholipid-binding proteins other than annexin II remains unclear. Because, in recent years, our research has focused on the identification

of endothelial autoantigens involved in different autoimmune diseases, studies based on the screening of endothelial cDNA expression libraries also identified vimentin as a new phospholipid-binding protein autoantigen in SN-APS [7]. Interestingly, in almost all the patients the positive result obtained by TLC assay was confirmed with the second result after at least 12 weeks; conversely, two patients negative with the first sample displayed aPL reactivity with the second sample. Of note, one of the last such cases was a 26-year-old female with MG-132 purchase SLE and proteinuria; histological evaluation of the kidney biopsy showed diffuse global lupus nephritis (class IV-G) associated with thrombotic microangiopathy suggestive of APS. Recently, it was demonstrated that aPL may exert

their pathogenic role by triggering a signal transduction pathway involving IRAK phosphorylation, NF-κB activation and translocation with consequent release of proinflammatory and procoagulant factors by endothelial and/or monocytic cells [18,20,25]. In order to verify the possible pathogenic role of the autoantibodies we demonstrate that purified IgG from sera of SN-APS patients induce IRAK serine phosphorylation with consequent NF-κB activation. Interestingly, we demonstrated that aCL as well as aLBPA were involved in this signalling pathway triggering, as these autoantibodies failed to induce many IRAK phosphorylation if they were

previously adsorbed with highly purified aCL or LBPA. Previous studies demonstrated that aPL induce monocyte and endothelial cell TF expression through the simultaneous activation of NF-κB-related proteins as well as aPL induce VCAM-1 on endothelial cells surface and that these effects are correlated with increased adhesion of leucocytes to endothelium [18,25,26]. According to these findings we demonstrate that IgG from SN-APS patients triggering resulted in the expression of VCAM-1, as well as release of TF from endothelial cells, which may contribute to the pathogenesis of thrombosis in patients with APS. Deep vein thrombosis, myocardial infarction and stroke are the major causes of morbidity and death among APS patients due to the high risk of recurrence; therefore, it is mandatory to identify among patients with suspected APS repeatedly negative for conventional aPL tests, those with a true APS to offer them long-term anti-coagulation, as widely recommended for secondary thromboprophylaxis in this disease [27,28].

However, these observations should be tempered by murine data sho

However, these observations should be tempered by murine data showing that IL-17 is produced from both CCR6- and CCR6+ Tregs at sites of disease (in this case, the CNS) [81]. In humans, the biological relevance of Treg to Th17 conversion

seen in vitro is unknown; however, human memory phenotype (CD45RO+) FoxP3+ Tregs isolated ex vivo have been shown very recently to secrete IL-17 and to express the Th17 transcription factor RORγt constitutively [85], suggesting that IL-17 production from Tregs also occurs in vivo. The reversal of the regulatory function of Tregs, and skewing of phenotype towards production of IL-17, a cytokine known to be important in human autoimmune diseases [60], may provide a link between the loss of regulation and high levels of IL-17 seen in some of these disorders. In addition, mice in which the IL-1 receptor antagonist gene has been silenced develop spontaneous autoimmune check details T cell-mediated arthritis, an IL-17-mediated condition [86,87], due to excessive IL-1 signalling [88]. These mice do not exhibit arthritis when kept germ-free, but rapidly develop pathological features when exposed to a single species of indigenous gut flora (Lactobacillus bifidus) or to signalling through TLRs [89]. The epidemiological association between infections and

the development of human autoimmune diseases could indicate a similar mechanism through altered Treg function and the promotion of IL-17, potentially also mediated through IL-1 or associated RAD001 mouse TLR signalling pathways. Demonstrations of the capacity of Tregs to convert to the Th17 lineage also suggests that infiltrating CD4+ cells bearing the phenotype of Tregs (CD4+CD25+FoxP3+) at sites of infection

[42] where IL-1β or IL-6 are highly expressed may not necessarily effect a suppressive function, but might instead participate in clearance of the inciting pathogen through conversion to the Th17 lineage. The stability of the Th17 phenotype in this model is an important Etofibrate consideration: given that Th17 cells generated from naive precursors are not stable either in vitro or in vivo[66–68], prolonged Treg-derived Th17 persistence at sites of inflammation may engender excessive tissue injury. Although this has not been addressed sufficiently in the literature, some available data suggest that restoration of suppressive function may be possible upon exposure to IL-2 [71]. In the context of concerted efforts to use expanded populations of Tregs for adoptive therapy in human inflammatory diseases, descriptions of Treg to Th17 conversion are important observations, as transition of adoptively transferred cells from an anti- to a proinflammatory lineage may exacerbate, rather than ameliorate, disease. Therefore, an understanding of the mechanisms underlying this conversion and methods to stabilize the Treg phenotype have become important aspects of Treg biology.

The UFFF was also preferred for its thinness and pliability (38%)

The UFFF was also preferred for its thinness and pliability (38%), reliable circulation due to perforators (18%), and the possibility of direct closure (49%) (Table 3). A.J. is a 67-year-old male who initially presented to the Otolaryngology-Head and Neck Surgery service at our institution with a left maxillary mass, biopsy positive for leiomyosarcoma. He was taken to the operating room with the Otolaryngology

service later that month where a left total maxillectomy and left orbital floor and orbital rim reconstruction were performed. this website A temporary obturator was placed and the orbital floor and rim were reconstructed with a titanium plate. Post-operatively, the patient received adjuvant chemotherapy and radiation. Approximately 6 months after surgery, the patient presented to Otolaryngology clinic with left facial cellulitis, ectropion, epiphora, and exposed

globe keratopathy. Silastic sheeting was seen to be protruding from the patient’s skin near the left medial canthus leaving a facial defect through which exposed hardware was visualized (Fig. 2). The patient was then seen by the Plastic Surgery service in consultation for reconstruction of the left maxillary defect. Consideration was Carfilzomib price given to free flap reconstruction using an anterolateral thigh versus vertical rectus abdominis myocutaneous flap versus RFFF. On exam, the patient was right-handed with positive modified Allen’s test findings suggesting the blood supply to the patient’s hands was radial artery-dominant (insufficient

collateral flow was noted through the ulnar artery with poor perfusion of the hand after release of the ulnar artery and observation for 15–20 seconds). The patient had a history of left wrist surgery resulting in a radial-based scar which precluded flap harvest from the left SPTLC1 side. The patient’s case was discussed in a joint conference with Otolaryngology, Oculoplastics, and Plastic Surgery. Given the patient’s responsibilities in caring for family members at home, it was felt that an UFFF would be the least likely flap to have complications and donor site morbidity and most likely to be closed primarily, thus allowing the patient to recover quickly and return to caring for his family. In addition, it was felt that the less hirsute UFFF would be preferable to a RFFF. The patient was taken to the operating room with the Otolaryngology and Plastic Surgery services; the exposed hardware was removed revealing a large, open cavity from the previous left total maxillectomy (Fig. 3). The remaining defect was reconstructed with a right UFFF (dimensions 3.5 × 10 cm, pedicle length 7 cm); perforating vessels of the ulnar artery were identified during UFFF harvest (Fig. 4).