Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 13 de 13
Filter
Add more filters










Publication year range
1.
Ann Surg Oncol ; 2024 Jul 13.
Article in English | MEDLINE | ID: mdl-39003380

ABSTRACT

BACKGROUND: The prognostic impact of genetic mutations for patients who undergo cytoreductive surgery (CRS) with hyperthermic intraperitoneal chemotherapy (HIPEC) of colorectal origin (CRC) is not well defined. OBJECTIVE: We aimed to describe the genetic classifications in an unsupervised fashion, and the outcomes of this patient population. METHODS: A retrospective, bi-institutional study was performed on patients who underwent CRS-HIPEC with targeted mutation data with a median follow-up time of 61 months. Functional link analysis was performed using STRING v11.5. Genes with similar functional significance were clustered using unsupervised k-means clustering. Chi-square, Kaplan-Meier, and the log-rank test were used for comparative statistics. RESULTS: Sixty-four patients with peritoneal carcinomatosis from CRC origin underwent CRS-HIPEC between 2007 and 2022 and genetic mutation data were extracted. We identified 19 unique altered genes, with KRAS (56%), TP53 (33%), and APC (22%) being the most commonly altered; 12.5% had co-altered KRAS/TP53. After creating an interactome map, k-means clustering revealed three functional clusters. Reactome Pathway analysis on three clusters showed unique pathways (1): Ras/FGFR3 signaling; (2) p53 signaling; and (3): NOTCH signaling. Seventy-one percent of patients in cluster 1 had KRAS mutations and a median overall survival of 52.3 months (p < 0.05). CONCLUSIONS: Patients with peritoneal carcinomatosis (PC) of CRC origin who underwent CRS-HIPEC and with tumors that harbored mutations in cluster 1 (Ras/FGFR3 signaling) had worse outcomes. Pathway disruption and a cluster-centric perspective may affect prognosis more than individual genetic alterations in patients with PC of CRC origin.

2.
Cureus ; 14(3): e23565, 2022 Mar.
Article in English | MEDLINE | ID: mdl-35495012

ABSTRACT

Background Traditionally, medical training programs have been viewed and ranked according to factors such as national prestige, research opportunities and productivity, faculty prominence, and alumni success. While these components are still instrumental in attracting high-level applicants, the rise of social media has encouraged applicants to look beyond the traditional media put forth by institutions and turn to sources such as program-specific, departmental, or institutional social media pages for more rapid updates. To date, little has been written on how to maximize residency program social media use to engage target audiences of current residents, current faculty, other physicians, and applicants. To better understand how certain types of content lead to increased engagement, we analyze the Instagram social media platform of the Mount Sinai Beth Israel Internal Medicine Residency Program to identify potential trends in interactions to ultimately enhance the resident experience and institutional standing. Methodology We reviewed 257 posts on our Instagram account and calculated engagement of each category relative to the post count based on a method we developed. We used Instagram analytics to better understand our account's reach. Results Posts highlighting social events had the highest engagement from the online community. Data also show that while the page is viewed by many people with the average medical student age, our page captures an even wider audience. Conclusions Instagram posts about the social element of our residency program generated statistically significant increased engagement. Institutions can use this strategy of focusing on the social aspects of the program to increase reputational scores within the medical and greater community.

3.
J Biol Chem ; 296: 100733, 2021.
Article in English | MEDLINE | ID: mdl-33957124

ABSTRACT

A disintegrin and metalloprotease 17 (ADAM17) is a cell-surface metalloprotease that serves as the principle sheddase for tumor necrosis factor α (TNFα), interleukin-6 receptor (IL-6R), and several ligands of the epidermal growth factor receptor (EGFR), regulating these crucial signaling pathways. ADAM17 activation requires its transmembrane domain, but not its cytoplasmic domain, and little is known about the role of this domain in vivo. To investigate, we used CRISPR-Cas9 to mutate the endogenous Adam17 locus in mice to produce a mutant ADAM17 lacking its cytoplasmic domain (Adam17Δcyto). Homozygous Adam17Δcyto animals were born at a Mendelian ratio and survived into adulthood with slightly wavy hair and curled whiskers, consistent with defects in ADAM17/EGFR signaling. At birth, Adam17Δcyto mice resembled Adam17-/- mice in that they had open eyes and enlarged semilunar heart valves, but they did not have bone growth plate defects. The deletion of the cytoplasmic domain resulted in strongly decreased ADAM17 protein levels in all tissues and cells examined, providing a likely cause for the hypomorphic phenotype. In functional assays, Adam17Δcyto mouse embryonic fibroblasts and bone-marrow-derived macrophages had strongly reduced ADAM17 activity, consistent with the reduced protein levels. Nevertheless, ADAM17Δcyto could be stimulated by PMA, a well-characterized posttranslational activator of ADAM17, corroborating that the cytoplasmic domain of endogenous ADAM17 is not required for its rapid response to PMA. Taken together, these results provide the first evidence that the cytoplasmic domain of ADAM17 plays a pivotal role in vivo in regulating ADAM17 levels and function.


Subject(s)
ADAM17 Protein/chemistry , ADAM17 Protein/metabolism , Cytoplasm/metabolism , ADAM17 Protein/genetics , Amino Acid Sequence , Animals , Base Sequence , CRISPR-Cas Systems , Female , Fibroblasts/metabolism , Male , Mice , Mice, Inbred C57BL , Phenotype , Protein Domains , Protein Stability , Sequence Deletion
4.
FASEB J ; 35(6): e21618, 2021 06.
Article in English | MEDLINE | ID: mdl-33982337

ABSTRACT

Tendons are dense connective tissues that transmit muscle forces to the skeleton. After adult injury, healing potential is generally poor and dominated by scar formation. Although the immune response is a key feature of healing, the specific immune cells and signals that drive tendon healing have not been fully defined. In particular, the immune regulators underlying tendon regeneration are almost completely unknown due to a paucity of tendon regeneration models. Using a mouse model of neonatal tendon regeneration, we screened for immune-related markers and identified upregulation of several genes associated with inflammation, macrophage chemotaxis, and TGFß signaling after injury. Depletion of macrophages using AP20187 treatment of MaFIA mice resulted in impaired functional healing, reduced cell proliferation, reduced ScxGFP+ neo-tendon formation, and altered tendon gene expression. Collectively, these results show that inflammation is a key component of neonatal tendon regeneration and demonstrate a requirement for macrophages in effective functional healing.


Subject(s)
Cell Proliferation , Inflammation/therapy , Macrophages/immunology , Regeneration , Tendon Injuries/therapy , Tenocytes/cytology , Wound Healing , Animals , Animals, Newborn , Disease Models, Animal , Female , Inflammation/immunology , Inflammation/pathology , Male , Mice , Tendon Injuries/immunology , Tendon Injuries/pathology , Tenocytes/physiology
5.
J Immunol ; 206(2): 302-309, 2021 01 15.
Article in English | MEDLINE | ID: mdl-33397744

ABSTRACT

Photosensitivity is a sensitivity to UV radiation (UVR) commonly found in systemic lupus erythematosus (SLE) patients who have cutaneous disease. Upon even ambient UVR exposure, patients can develop inflammatory skin lesions that can reduce the quality of life. Additionally, UVR-exposed skin lesions can be associated with systemic disease flares marked by rising autoantibody titers and worsening kidney disease. Why SLE patients are photosensitive and how skin sensitivity leads to systemic disease flares are not well understood, and treatment options are limited. In recent years, the importance of immune cell-stromal interactions in tissue function and maintenance is being increasingly recognized. In this review, we discuss SLE as an anatomic circuit and review recent findings in the pathogenesis of photosensitivity with a focus on immune cell-stromal circuitry in tissue health and disease.


Subject(s)
Lupus Erythematosus, Systemic/immunology , Photosensitivity Disorders/immunology , Skin/pathology , Animals , Autoantibodies/metabolism , Cell Communication , Humans , Immunity, Cellular
6.
J Appl Physiol (1985) ; 128(3): 473-482, 2020 03 01.
Article in English | MEDLINE | ID: mdl-31944888

ABSTRACT

Tendon injuries are a common clinical condition with limited treatment options. The cellular components of the innate immune system, such as neutrophils and macrophages, have been studied in tendon injuries. However, the adaptive immune system, comprising specialized lymphocytes, plays an important role in orchestrating the healing of numerous tissues, but less is known about these cells in tendon healing. To gain a greater understanding of the biological processes that regulate tendon healing, we determined how the cellular components of the adaptive and innate immune system respond to a tendon injury using two-month-old male mice. We observed that lymphatic vasculature is present in the epitenon and superficial regions of Achilles tendons, and that the lymphatics drain into the popliteal lymph node. We then created an acute Achilles tenotomy followed by repair, and collected tendons and popliteal lymph nodes 1, 2, and 4 wk after injury. Tendon injury resulted in a robust adaptive immune cell response that followed an initial innate immune cell response in tendons and lymph nodes. Monocytes, neutrophils, and macrophages initially accumulated at 1 wk after injury in tendons, while dendritic cells and CD4+ T cells peaked at 2 wk after injury. B cells and CD8+ T cells progressively increased over time. In parallel, immune cells of the popliteal lymph node demonstrated a similarly coordinated response to the injury. These results suggest that there is an adaptive immune response to tendon injury, and adaptive immune cells may play a role in regulating tendon healing.NEW & NOTEWORTHY While the innate immune system, consisting of macrophages and related hematopoietic cells, has been studied in tendon injury, less is known about the adaptive immune system. Using a mouse model of Achilles tendon tenotomy and repair, we observed an adaptive immune cell response, consisting of CD4+ and CD8+ T cells, and B cells, which occur through 4 wk after tendon injury. This response appeared to be coordinated by the draining popliteal lymph node.


Subject(s)
Achilles Tendon , Tendon Injuries , CD8-Positive T-Lymphocytes , Humans , Immunity, Innate , Lymph Nodes , Male
7.
Clin Orthop Relat Res ; 477(11): 2588-2598, 2019 Nov.
Article in English | MEDLINE | ID: mdl-31283731

ABSTRACT

BACKGROUND: Periprosthetic joint infection (PJI) is one of the most devastating complications of total joint arthroplasty. Given the mortality and morbidity associated with PJI and the challenges in treating it, there has been increased interest in risk factors that can be modified before surgery. In this study, we used a novel mouse model to consider the role of the gut microbiome as a risk factor for PJI. QUESTIONS/PURPOSES: (1) Does the state of the gut microbiota before surgery influence the likelihood of developing an established infection in a mouse model of PJI? (2) How does the state of the gut microbiota before surgery influence the local and systemic response to the presence of an established infection in a mouse model of PJI? METHODS: Male C57Bl/6 mice were divided into two groups: those with modified microbiome [INCREMENT]microbiome (n = 40) and untreated mice (n = 42). In [INCREMENT]microbiome mice, the gut flora were modified using oral neomycin and ampicillin from 4 weeks to 16 weeks of age. Mice received a titanium tibial implant to mimic a joint implant and a local inoculation of Staphylococcus aureus in the synovial space (10 colony forming units [CFUs]). The proportion of animals developing an established infection in each group was determined by CFU count. The local and systemic response to established infection was determined using CFU counts in surrounding joint tissues, analysis of gait, radiographs, body weight, serum markers of inflammation, and immune cell profiles and was compared with animals that received the inoculation but resisted infection. RESULTS: A greater proportion of animals with disrupted gut microbiota had infection (29 of 40 [73%]) than did untreated animals (21 of 42 [50%]; odds ratio, 2.63, 95% CI, 1.04-6.61; p = 0.035). The immune response to established infection in mice with altered microbiota was muted; serum amyloid A, a marker of systemic infection in mice, was greater than in mice with disrupted gut microbiota with infection (689 µg/dL; range, 68-2437 µg/dL, p < 0.05); infection associated increases in monocytes and neutrophils in the spleen and local lymph node in untreated mice but not were not observed in mice with disrupted gut microbiota. CONCLUSIONS: The findings from this in vivo mouse model suggest that the gut microbiota may influence susceptibility to PJI. CLINICAL RELEVANCE: These preclinical findings support the idea that the state of the gut microbiome before surgery may influence the development of PJI and justify further preclinical and clinical studies to develop appropriate microbiome-based interventions.


Subject(s)
Gastrointestinal Microbiome/physiology , Joint Prosthesis/adverse effects , Prosthesis-Related Infections/etiology , Staphylococcal Infections/etiology , Staphylococcus aureus , Tibia/surgery , Animals , Disease Models, Animal , Mice
8.
Front Immunol ; 10: 519, 2019.
Article in English | MEDLINE | ID: mdl-30949174

ABSTRACT

Lymphatic vessels are critical for clearing fluid and inflammatory cells from inflamed tissues and also have roles in immune tolerance. Given the functional association of the lymphatics with the immune system, lymphatic dysfunction may contribute to the pathophysiology of rheumatic autoimmune diseases. Here we review the current understanding of the role of lymphatics in the autoimmune diseases rheumatoid arthritis, scleroderma, lupus, and dermatomyositis and consider the possibility that manual therapies such as massage and acupuncture may be useful in improving lymphatic function in autoimmune diseases.


Subject(s)
Arthritis, Rheumatoid/immunology , Lymphatic Vessels/immunology , Animals , Arthritis, Rheumatoid/pathology , Humans , Lymphatic Vessels/pathology
11.
Clin Chim Acta ; 361(1-2): 199-205, 2005 Nov.
Article in English | MEDLINE | ID: mdl-15996649

ABSTRACT

BACKGROUND: Allergen-specific IgE (sIgE) measurements are used to help identify causative allergenic agents and to determine the degree of sensitization to facilitate treatment decisions. We examined the performance of a new third-generation chemiluminescent enzyme immunoassay for allergen-specific IgE (sIgE) on the continuous random access Immulite 2000 system. METHODS: Detection limit and dilutional linearity were determined. Within-run and total precision were assessed according to the NCCLS EP5 guideline. Interlaboratory comparison of the new Immulite 2000 third-generation allergen-specific IgE assay was performed, as well as intermethod comparison against the Pharmacia FEIA, a second-generation assay. RESULTS: The detection limit was <0.1 kU/l. Dilutional linearity held from 100 down to 0.2 kU/l. Regression analysis of the interlaboratory comparison results yielded: Immulite 2000(Laboratory 1)=1.07 Immulite 2000(Laboratory 2)+0.18 kU/l; r=0.98, n=3588 results. Intermethod comparison showed the following: Immulite 2000=0.83 (Pharmacia FEIA)+0.42 kU/l; r=0.79, n=512 results. Bland-Altman analysis of the interlaboratory and intermethod comparisons indicated no systematic bias. CONCLUSIONS: We confirmed the reported performance characteristics of the third-generation sIgE assay and found reasonably good interlaboratory and intermethod agreement. The extended range capability of the third-generation assay provides a new tool for investigating cutoffs and trends in childhood allergy disease progression at concentrations <0.35 kU/l.


Subject(s)
Allergens/immunology , Immunoassay/standards , Immunoglobulin E/blood , Immunoglobulin E/immunology , Antibody Specificity , Dermatitis, Atopic/blood , Dermatitis, Atopic/immunology , Humans , Reproducibility of Results
12.
Ann Clin Lab Sci ; 34(1): 67-74, 2004.
Article in English | MEDLINE | ID: mdl-15038670

ABSTRACT

In vitro determination of allergen-specific IgE (sIgE) represents an important aid in the diagnosis and treatment of allergy. Improvements in laboratory methodology--the development of second, and now third generation assays for sIgE--have brought about major advances in speed, convenience, performance, and in the standards for judging performance. In this study, following the NCCLS I/LA20-A guidelines, we evaluated the analytical performance of a quantitative chemiluminescent enzyme immunoassay for sIgE using the continuous random access IMMULITE 2000 system. Defining features of this "third generation" sIgE assay include a true zero calibrator with a detection limit and functional sensitivity of 0.1 and 0.2 kU/L, respectively. The use of liquid allergens allows for complete automation, fast binding kinetics between IgE and the natural allergenic protein conformations, and a time-to-first-result of 65 min. Stable reagents and the low nonspecific signal associated with the liquid allergens and centrifugal wash technique permit extension of the measuring range to 0.1-100 kU/L, based on lot-specific, factory-calibrated master curves standardized to the WHO 75/502 reference standard. The assay demonstrated good precision and linearity over its measuring range. Relative to a first generation RIA (mRAST, from Hycor), clinical sensitivity, specificity, and concordance were 88%, 92%, and 90%, respectively (n = 812). Quantitative comparisons to a second generation assay yielded a linear regression relationship of IMMULITE 2000 = 0.99 (Pharmacia FEIA) + 1.99 kU/L, r = 0.859 (n = 169).


Subject(s)
Allergens/immunology , Immunoenzyme Techniques/methods , Immunoglobulin E/analysis , Humans , Immunoenzyme Techniques/instrumentation , Immunoglobulin E/immunology
SELECTION OF CITATIONS
SEARCH DETAIL
...