Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 28
Filter
1.
Animals (Basel) ; 14(3)2024 Jan 31.
Article in English | MEDLINE | ID: mdl-38338104

ABSTRACT

Digital dermatitis is a disease of the digital skin and causes lameness and welfare problems in dairy cattle. This study assessed the local and systemic inflammatory responses of cows with different digital dermatitis lesions and compared macroscopical and histological findings. Cow feet (n = 104) were evaluated macroscopically and skin biopsies histologically. Serum samples were analyzed for acute phase proteins (serum amyloid A and haptoglobin) and pro-inflammatory cytokines (interleukin-1 beta, interleukin-6, and tumor necrosis factor-alpha). Cows with macroscopically graded active lesions (p = 0.028) and non-active lesions (p = 0.008) had higher interleukin-1 beta levels in their serum compared to healthy cows. Interleukin-1 beta serum concentrations were also higher (p = 0.042) when comparing lesions with necrosis to lesions without necrosis. There was no difference when other cytokine or acute phase protein concentrations in healthy cows were compared to those in cows with different digital dermatitis lesions. A novel histopathological grading was developed based on the chronicity of the lesions and presence of necrosis and ulceration. The presence and number of spirochetes were graded separately. In the most severe chronic lesions, there was marked epidermal hyperplasia and hyperkeratosis with necrosis, deep ulceration, and suppurative inflammation. Spirochetes were found only in samples from necrotic lesions. This study established that digital dermatitis activates proinflammatory cytokines. However, this did not initiate the release of acute phase proteins from the liver. A histopathological grading that takes into account the age and severity of the lesions and presence of spirochetes was developed to better understand the progression of the disease. It is proposed that necrosis of the skin is a result of ischemic necrosis following reduced blood flow in the dermal papillae due to pressure and shear stress caused by thickened epidermis, and that the spirochetes are secondary invaders following tissue necrosis.

2.
Front Immunol ; 14: 1171083, 2023.
Article in English | MEDLINE | ID: mdl-37475863

ABSTRACT

Pancreatic ductal adenocarcinoma (PDAC) is a highly treatment-resistant cancer. Currently, the only curative treatment for PDAC is surgery, but most patients are diagnosed with metastatic disease and thus outside the scope of surgery. The majority of metastatic patients receive chemotherapy, but responses are limited. New therapeutics are thus urgently needed for PDAC. One major limitation in treating PDAC has been the highly immunosuppressive tumor microenvironment (TME) which inhibits anti-cancer immune responses. We have constructed an oncolytic adenovirus coding for a variant the interleukin 2 molecule, Ad5/3-E2F-d24-vIL2 (also known as TILT-452, and "vIL-2 virus"), with preferential binding to IL-2 receptors on the surface of effector lymphocytes over T regulatory cells (T regs). In the present study this virus was evaluated in combination with nab-paclitaxel and gemcitabine chemotherapy in Panc02 mouse model. Ad5/3-E2F-d24-vIL2 showed marked PDAC cell killing in vitro, alongside induction of mitotic slippage and immunogenic cell death in PDAC cell lines, when combined with chemotherapy. Increased survival was seen in vivo with 80% of animals surviving long term, when compared to chemotherapy alone. Moreover, combination therapy mediated enhanced tumor growth control, without observable toxicities in internal organs or external features. Survival and tumor control benefits were associated with activation of tumor infiltrating immune cells, downregulation of inhibitory signals, change in fibroblast populations in the tumors and changes in intratumoral cytokines, with increased chemokine amounts (CCL2, CCL3, CCL4) and anti-tumor cytokines (IFN-γ and TNFα). Furthermore, vIL-2 virus in combination with chemotherapy efficiently induced tumor protection upon rechallenge, that was extended to a previously non-encountered cancer cell line. In conclusion, Ad5/3-E2F-d24-vIL2 is a promising immunotherapy candidate when combined with nab-paclitaxel and gemcitabine.


Subject(s)
Adenoviridae Infections , Carcinoma, Pancreatic Ductal , Pancreatic Neoplasms , Animals , Mice , Adenoviridae , Cytokines/therapeutic use , Interleukin-2/genetics , Interleukin-2/pharmacology , Interleukin-2/therapeutic use , Cell Line, Tumor , Carcinoma, Pancreatic Ductal/therapy , Carcinoma, Pancreatic Ductal/drug therapy , Gemcitabine , Lymphocytes/pathology , Fibroblasts/pathology , Tumor Microenvironment , Pancreatic Neoplasms
3.
Genes (Basel) ; 14(5)2023 04 27.
Article in English | MEDLINE | ID: mdl-37239348

ABSTRACT

Sudden cardiac death in the young (SCDY) is a devastating event that often has an underlying genetic basis. Manchester Terrier dogs offer a naturally occurring model of SCDY, with sudden death of puppies as the manifestation of an inherited dilated cardiomyopathy (DCM). We performed a genome-wide association study for SCDY/DCM in Manchester Terrier dogs and identified a susceptibility locus harboring the cardiac ATP-sensitive potassium channel gene ABCC9. Sanger sequencing revealed an ABCC9 p.R1186Q variant present in a homozygous state in all SCDY/DCM-affected dogs (n = 26). None of the controls genotyped (n = 398) were homozygous for the variant, but 69 were heterozygous carriers, consistent with autosomal recessive inheritance with complete penetrance (p = 4 × 10-42 for the association of homozygosity for ABCC9 p.R1186Q with SCDY/DCM). This variant exists at low frequency in human populations (rs776973456) with clinical significance previously deemed uncertain. The results of this study further the evidence that ABCC9 is a susceptibility gene for SCDY/DCM and highlight the potential application of dog models to predict the clinical significance of human variants.


Subject(s)
Cardiomyopathy, Dilated , Death, Sudden, Cardiac , Dog Diseases , Sulfonylurea Receptors , Animals , Dogs , Cardiomyopathy, Dilated/genetics , Cardiomyopathy, Dilated/veterinary , Death, Sudden, Cardiac/etiology , Death, Sudden, Cardiac/veterinary , Genome-Wide Association Study , Genotype , Mutation, Missense , Sulfonylurea Receptors/genetics , Dog Diseases/genetics
4.
Front Immunol ; 14: 1060540, 2023.
Article in English | MEDLINE | ID: mdl-36817448

ABSTRACT

Introduction: Immune checkpoint inhibitors (ICIs) have revolutionized the treatment of cancer, but preclinical testing of hypotheses such as combination therapies has been complicated, in part due to species incompatibility issues. For example, one of few known permissive animal models for oncolytic adenoviruses is the Syrian hamster, for which an ICI, mainly an anti-PD-L1 monoclonal antibody (mAb) was not previously available. In this study, we developed an anti-Syrian hamster PD-L1 mAb to enable the evaluation of safety and efficacy, when combining anti-PD-L1 with an oncolytic adenovirus encoding tumour necrosis factor alpha (TNFα) and interleukin-2 (IL-2) (Ad5/3-E2F-D24-hTNFα-IRES-hIL-2 or TILT-123). Methods: Recombinant Syrian hamster PD-L1 was expressed and mice immunized for mAb formation using hybridoma technology. Clonal selection through binding and functional studies in vitro, in silico and in vivo identified anti-PD-L1 clone 11B12-1 as the primary mAb candidate for immunotherapy modelling. The oncolytic virus (OV) and ICI combination approach was then evaluated using 11B12-1 and TILT-123 in a Syrian hamster model of pancreatic ductal adenocarcinoma (PDAC). Results: Supernatants from hybridoma parent subclone 11B12B4 provided the highest positive PD-L1 signal, on Syrian hamster PBMCs and three cancer cell lines (HT100, HapT1 and HCPC1). In vitro co-cultures revealed superior immune modulated profiles of cell line matched HT100 tumour infiltrating lymphocytes when using subclones of 7G2, 11B12 and 12F1. Epitope binning and epitope prediction using AlphaFold2 and ColabFold revealed two distinct functional epitopes for clone 11B12-1 and 12F1-1. Treatment of Syrian hamsters bearing HapT1 tumours, with 11B12-1 induced significantly better (p<0.05) tumour growth control than isotype control by day 12. 12F1-1 did not induce significant tumour growth control. The combination of 11B12-1 with oncolytic adenovirus TILT-123 improved tumour growth control further, when compared to monotherapy (p<0.05) by day 26. Conclusions: Novel Syrian hamster anti-PD-L1 clone 11B12-1 induces tumour growth control in a hamster model of PDAC. Combining 11B12-1 with oncolytic adenovirus TILT-123 improves tumour growth control further and demonstrates good safety and toxicity profiles.


Subject(s)
Carcinoma, Pancreatic Ductal , Oncolytic Viruses , Pancreatic Neoplasms , Cricetinae , Animals , Mice , Mesocricetus , Immune Checkpoint Inhibitors , Adenoviridae , Pancreatic Neoplasms/therapy , Immunotherapy , Antibodies, Monoclonal , Virus Replication , Pancreatic Neoplasms
5.
Mol Ther Oncolytics ; 28: 59-73, 2023 Mar 16.
Article in English | MEDLINE | ID: mdl-36699617

ABSTRACT

Immunotherapy with bispecific T cell engagers has shown efficacy in patients with hematologic malignancies and uveal melanoma. Antitumor effects of bispecific T cell engagers in most solid tumors are limited due to their short serum half-life and insufficient tumor concentration. We designed a novel serotype 5/3 oncolytic adenovirus encoding a human mucin1 antibody and the human CD3 receptor, Ad5/3-E2F-d24-aMUC1aCD3 (TILT-321). TILT-321 is engineered to replicate only in cancer cells, leading to a high concentration of the aMUC1aCD3 molecule in the tumor microenvironment. Infection and cell viability assays were performed to determine the oncolytic potential of the novel construct. The functionality of the virus-derived aMUC1aCD3 was evaluated in vitro. When TILT-321 was combined with allogeneic T cells, rapid tumor cell lysis was observed. TILT-321-infected cells secreted functional aMUC1aCD3, as shown by increased T cell activity and its binding to MUC1 and CD3. In vivo, TILT-321 treatment led to effective antitumor efficacy mediated by increased intratumoral T cell activity in an A549 and patient-derived ovarian cancer xenograft mouse model humanized with peripheral blood mononuclear cells (PBMC). This study provides a proof of concept for an effective strategy to overcome the key limitations of recombinant bispecific T cell engager delivery for solid tumor treatment.

6.
Oncoimmunology ; 11(1): 2028960, 2022.
Article in English | MEDLINE | ID: mdl-35083096

ABSTRACT

Intratumoral immunotherapies are entering clinical use but concerns remain regarding their effects on non-injected tumors. Here, we studied the impact of local treatment with an adenovirus coding for TNFa and IL-2 on systemic antitumor response in animals receiving aPD-1 (anti-programmed cell death protein 1) therapy. Using bilateral murine melanoma models, we tested systemic tumor response to combined therapy with anti-PD-1 and an adenovirus coding for TNFa and IL-2 ("virus"). Virus was given intratumorally (to one of the two tumors only) and aPD-1 monoclonal antibody systemically. We evaluated both tumors' response to treatment, overall survival, metastasis development, and immunological mechanisms involved with response. Consistent tumor control was observed in both injected and non-injected tumors, including complete response in all treated animals receiving aPD-1+ virus therapy. Mechanistically, virus injections enabled potent effector lymphocyte response locally, with systemic effects in non-injected tumors facilitated by aPD-1 treatment. Moreover, adenovirus therapy demonstrated immunological memory formation. Virus therapy was effective in preventing metastasis development. Local treatment with TNFa and IL-2 coding adenovirus enhanced systemic response to aPD-1 therapy, by re-shaping the microenvironment of both injected and non-injected tumors. Therefore, our pre-clinical data support the rationale for a trial utilizing a combination of aPD-1 plus virus for the treatment of human cancer.


Subject(s)
Adenoviridae Infections , Melanoma , Oncolytic Virotherapy , Adenoviridae/genetics , Animals , Immunotherapy , Interleukin-2 , Melanoma/therapy , Mice , Tumor Microenvironment
7.
J Equine Vet Sci ; 104: 103694, 2021 09.
Article in English | MEDLINE | ID: mdl-34416991

ABSTRACT

Exercise-associated sudden deaths (EASDs) are deaths occurring unexpectedly during or immediately after exercise. Sudden cardiac death (SCD) is one cause of EASD. Cardiac arrhythmias caused by genetic variants have been linked to SCD in humans. We hypothesize that genetic variants may be associated with SCD in animals, including horses. Genetic variants are transmitted to offspring and their frequency might increase within a family. Therefore, the frequency of such variants might increase with the inbreeding factor. Higher inbreeding could have a negative impact on racing performance. Pedigree data and career earnings from racehorses diagnosed with SCD between 2002 and 2017 were compared using non-parametric tests with 1) control horses that died due to catastrophic musculoskeletal injuries and 2) horses that raced during the same period without reported problems. Diagnosis of SCD was based on necropsy reports, including macroscopic and microscopic examinations. Death was registered in the study period for 61 horses. Eleven of these horses were excluded due to missing autopsy reports. In 25 cases, the diagnosis remained unknown and death was possibly caused by cardiac arrhythmia, in two cases cardiac disease was identified, in seven cases a rupture of a major vessel had occurred. In addition, 16 horses died or were euthanized due to severe musculoskeletal injuries. No significant differences in inbreeding coefficients or in career earnings were found between the groups or between horses with EASD compared with other horses racing during the same period. The study provides no evidence for increased inbreeding factor in Finnish racehorses with SCD.


Subject(s)
Death, Sudden, Cardiac , Horse Diseases , Physical Conditioning, Animal , Animals , Death, Sudden, Cardiac/epidemiology , Death, Sudden, Cardiac/veterinary , Euthanasia, Animal , Finland/epidemiology , Horse Diseases/genetics , Horses , Humans , Pedigree
8.
Front Immunol ; 12: 674400, 2021.
Article in English | MEDLINE | ID: mdl-34084172

ABSTRACT

The notion of developing variants of the classic interleukin 2 (IL-2) cytokine has emerged from the limitations observed with the systemic use of human IL-2 in the clinic: severe adverse events accompanied by low therapeutic response rate in treated patients. Modifications made in the IL-2 receptor-binding structure leads to preferential binding of IL-2 variant cytokine to receptors on effector anti-tumor lymphocytes over T regulatory (TReg) cells. Because of their inherent immunogenicity, oncolytic adenoviruses are useful for expression of immunomodulatory molecules in tumors, for induction of a pro-inflammatory state in the tumor microenvironment. In the present study, we constructed an adenovirus coding for an IL-2 variant (vIL-2) protein, Ad5/3-E2F-d24-vIL2. Functionality of the new virus was tested in vitro, and anti-tumor efficacy and mechanism of action studies were performed in immunocompetent hamsters bearing pancreatic tumors. Ad5/3-E2F-d24-vIL2 treatment elicited efficient anti-tumor response, with 62.5% monotherapy complete response. Moreover, it promoted substantial repression of genes associated with myeloid cells mediated immunosuppression (CD11b, ARG1, CD206). This was seen in conjunction with upregulation of genes associated with tumor-infiltrating lymphocyte (TIL) cytotoxicity (CD3G, SAP, PRF1, GZMM and GZMK). In summary, Ad5/3-E2F-d24-vIL2 demonstrates therapeutic potential by counteracting immunosuppression and in efficiently coordinating lymphocytes mediated anti-tumor response in immunosuppressive tumors. Thus, Ad5/3-E2F-d24-vIL2 is a promising candidate for translation into clinical trials in human immunosuppressive solid tumors.


Subject(s)
Adenoviridae , Genetic Vectors , Interleukin-2/immunology , Pancreatic Neoplasms/immunology , Tumor Microenvironment/immunology , Animals , Cricetinae , Humans , Interleukin-2/genetics , Male , Mesocricetus , Oncolytic Viruses , Recombinant Proteins/genetics , Recombinant Proteins/immunology
9.
Hum Genet ; 140(11): 1593-1609, 2021 Nov.
Article in English | MEDLINE | ID: mdl-33835239

ABSTRACT

We investigated the clinical, genetic, and pathological characteristics of a previously unknown severe juvenile brain disorder in several litters of Parson Russel Terriers. The disease started with epileptic seizures at 6-12 weeks of age and progressed rapidly to status epilepticus and death or euthanasia. Histopathological changes at autopsy were restricted to the brain. There was severe acute neuronal degeneration and necrosis diffusely affecting the grey matter throughout the brain with extensive intraneuronal mitochondrial crowding and accumulation of amyloid-ß (Aß). Combined homozygosity mapping and genome sequencing revealed an in-frame 6-bp deletion in the nuclear-encoded pitrilysin metallopeptidase 1 (PITRM1) encoding for a mitochondrial protease involved in mitochondrial targeting sequence processing and degradation. The 6-bp deletion results in the loss of two amino acid residues in the N-terminal part of PITRM1, potentially affecting protein folding and function. Assessment of the mitochondrial function in the affected brain tissue showed a significant deficiency in respiratory chain function. The functional consequences of the mutation were modeled in yeast and showed impaired growth in permissive conditions and an impaired respiration capacity. Loss-of-function variants in human PITRM1 result in a childhood-onset progressive amyloidotic neurological syndrome characterized by spinocerebellar ataxia with behavioral, psychiatric and cognitive abnormalities. Homozygous Pitrm1-knockout mice are embryonic lethal, while heterozygotes show a progressive, neurodegenerative phenotype characterized by impairment in motor coordination and Aß deposits. Our study describes a novel early-onset PITRM1-related neurodegenerative canine brain disorder with mitochondrial dysfunction, Aß accumulation, and lethal epilepsy. The findings highlight the essential role of PITRM1 in neuronal survival and strengthen the connection between mitochondrial dysfunction and neurodegeneration.


Subject(s)
Dog Diseases/genetics , Epilepsy/veterinary , Metalloendopeptidases/genetics , Mitochondria/metabolism , Neurodegenerative Diseases/veterinary , Amyloid beta-Peptides/metabolism , Animals , Brain/enzymology , Brain/metabolism , Brain/pathology , Dog Diseases/pathology , Dogs , Epilepsy/genetics , Female , Male , Metalloendopeptidases/chemistry , Metalloendopeptidases/metabolism , Neurodegenerative Diseases/genetics , Neurodegenerative Diseases/pathology , Oxygen Consumption , Pedigree , Saccharomyces cerevisiae/genetics , Saccharomyces cerevisiae/growth & development , Saccharomyces cerevisiae/metabolism
10.
Cells ; 10(2)2021 01 27.
Article in English | MEDLINE | ID: mdl-33513935

ABSTRACT

Oncolytic viruses provide a biologically multi-faceted treatment option for patients who cannot be cured with currently available treatment options. We constructed an oncolytic adenovirus, TILT-123, to support T-cell therapies and immune checkpoint inhibitors in solid tumors. Adenoviruses are immunogenic by nature, are easy to produce in large quantities, and can carry relatively large transgenes. They are the most commonly used gene therapy vectors and are well tolerated in patients. TILT-123 expresses two potent cytokines, tumor necrosis factor alpha and interleukin-2, to stimulate especially the T-cell compartment in the tumor microenvironment. Before entering clinical studies, the safety and biodistribution of TILT-123 was studied in Syrian hamsters and in mice. The results show that TILT-123 is safe in animals as monotherapy and in combination with an immune checkpoint inhibitor anti-PD-1. The virus treatment induces acute changes in circulating immune cell compartments, but the levels return to normal by the middle of the treatment period. The virus is rapidly cleared from healthy tissues, and it does not cause damage to vital organs. The results support the initiation of a phase 1 dose-escalation trial, where melanoma patients receiving a tumor-infiltrating lymphocyte therapy are treated with TILT-123 (NCT04217473).


Subject(s)
Adenoviridae/metabolism , Cytokines/metabolism , Immune Checkpoint Inhibitors/pharmacology , Oncolytic Viruses/metabolism , Programmed Cell Death 1 Receptor/antagonists & inhibitors , Animals , Antibodies, Neutralizing/metabolism , Cell Line, Tumor , Cricetinae , Female , Injections , Male , Mice , Neoplasms/immunology , Neoplasms/pathology , Organ Specificity , Programmed Cell Death 1 Receptor/metabolism , Tissue Distribution , Transgenes , Virus Replication/physiology , Xenograft Model Antitumor Assays
11.
Oncoimmunology ; 9(1): 1761229, 2020 05 22.
Article in English | MEDLINE | ID: mdl-32923123

ABSTRACT

Checkpoint inhibitors have revolutionized cancer therapy and validated immunotherapy as an approach. Unfortunately, responses are seen in a minority of patients. Our objective is to use engineered adenoviruses designed to increase lymphocyte trafficking and cytokine production at the tumor, to assess if they increase the response rate to checkpoint inhibition, as these features have been regarded as predictive for the responses. When Ad5/3-E2F-d24-hTNFa-IRES-hIL2 (an oncolytic adenovirus coding for TNFa and IL-2, also known as TILT-123) and checkpoint inhibitors were used together in fresh urological tumor histocultures, a significant shift toward immune activity (not only tumor necrosis alpha and interleukin-2 but also interferon gamma and granzyme B) and increased T-cell trafficking signals (CXCL10) was observed. In vivo, our viruses enabled an anti-PD-L1 (a checkpoint inhibitor) delivering complete responses in all the treated animals (hazard ratios versus anti-PD-L1 alone 0.057 [0.007; 0.451] or virotherapy alone 0.067 [0.011; 0.415]). To conclude, when an engineered oncolytic adenovirus was utilized to modify the tumor microenvironment towards what meta-analyses have pointed as predictive markers for checkpoint inhibitory therapy, the response to them increased synergistically. Of note, key findings were confirmed in fresh patient-derived tumor explants.


Subject(s)
Oncolytic Virotherapy , Oncolytic Viruses , Adenoviridae/genetics , Animals , B7-H1 Antigen/genetics , Humans , Oncolytic Viruses/genetics , Tumor Microenvironment
12.
Mol Ther Oncolytics ; 17: 47-60, 2020 Jun 26.
Article in English | MEDLINE | ID: mdl-32322662

ABSTRACT

Despite some promising results, the majority of patients do not benefit from T cell therapies, as tumors prevent T cells from entering the tumor, shut down their activity, or downregulate key antigens. Due to their nature and mechanism of action, oncolytic viruses have features that can help overcome many of the barriers currently facing T cell therapies of solid tumors. This study aims to understand how four different oncolytic viruses (adenovirus, vaccinia virus, herpes simplex virus, and reovirus) perform in that task. For that purpose, an immunocompetent in vivo tumor model featuring adoptive tumor-infiltrating lymphocyte (TIL) therapy was used. Tumor growth control (p < 0.001) and survival analyses suggest that adenovirus was most effective in enabling T cell therapy. The complete response rate was 62% for TILs + adenovirus versus 17.5% for TILs + PBS. Of note, TIL biodistribution did not explain efficacy differences between viruses. Instead, immunostimulatory shifts in the tumor microenvironment mirrored efficacy results. Overall, the use of oncolytic viruses can improve the utility of T cell therapies, and additional virus engineering by arming with transgenes can provide further antitumor effects. This phenomenon was seen when an unarmed oncolytic adenovirus was compared to Ad5/3-E2F-d24-hTNFa-IRES-hIL2 (TILT-123). A clinical trial is ongoing, where patients receiving TIL treatment also receive TILT-123 (ClinicalTrials.gov: NCT04217473).

13.
PLoS Genet ; 16(3): e1008651, 2020 03.
Article in English | MEDLINE | ID: mdl-32150563

ABSTRACT

Neonatal interstitial lung diseases due to abnormal surfactant biogenesis are rare in humans and have never been reported as a spontaneous disorder in animals. We describe here a novel lung disorder in Airedale Terrier (AT) dogs with clinical symptoms and pathology similar to the most severe neonatal forms of human surfactant deficiency. Lethal hypoxic respiratory distress and failure occurred within the first days or weeks of life in the affected puppies. Transmission electron microscopy of the affected lungs revealed maturation arrest in the formation of lamellar bodies (LBs) in the alveolar epithelial type II (AECII) cells. The secretory organelles were small and contained fewer lamellae, often in combination with small vesicles surrounded by an occasionally disrupted common limiting membrane. A combined approach of genome-wide association study and whole exome sequencing identified a recessive variant, c.1159G>A, p.(E387K), in LAMP3, a limiting membrane protein of the cytoplasmic surfactant organelles in AECII cells. The substitution resides in the LAMP domain adjacent to a conserved disulfide bond. In summary, this study describes a novel interstitial lung disease in dogs, identifies a new candidate gene for human surfactant dysfunction and brings important insights into the essential role of LAMP3 in the process of the LB formation.


Subject(s)
Lung Diseases, Interstitial/genetics , Lysosomal-Associated Membrane Protein 3/genetics , ATP-Binding Cassette Transporters/genetics , Animals , Dogs , Female , Genome-Wide Association Study , Lung/metabolism , Lung Diseases, Interstitial/physiopathology , Lysosomal-Associated Membrane Protein 3/metabolism , Lysosomal Membrane Proteins/genetics , Lysosomal Membrane Proteins/metabolism , Male , Microscopy, Electron, Transmission , Mutation, Missense , Organelles/metabolism , Pulmonary Alveoli/metabolism , Pulmonary Surfactants , Secretory Vesicles/metabolism
14.
J Vet Intern Med ; 33(3): 1376-1383, 2019 May.
Article in English | MEDLINE | ID: mdl-30844093

ABSTRACT

BACKGROUND: There is a successful outcome after surgical management of spinal arachnoid diverticula (SAD) in up to 82% of cases. HYPOTHESIS/OBJECTIVES: We hypothesized that Pugs have favorable short-term and poor long-term prognosis after surgical treatment of thoracolumbar SAD. The aim of the present investigation was to describe clinical findings, short- and long-term outcomes, and follow-up magnetic resonance imaging (MRI) findings in Pugs with thoracolumbar SAD. ANIMALS: Twenty-five client owned Pugs with 12-month follow-up information after surgical treatment of thoracolumbar SAD. METHODS: Multicenter retrospective case series. All medical records were searched for Pugs diagnosed with SAD. Data regarding signalment, history, surgical procedure, outcome, histopathology, and follow-up MRI results were extracted. RESULTS: Mean age at presentation was 7.32 (range 2-11) years, 80% were males. Short-term outcome was available in 25 dogs, and improvement was confirmed in 80% of dogs. Long-term outcome was available in 21 dogs, and deterioration was confirmed in 86% of cases, with late-onset recurrence of clinical signs after initial postsurgical improvement affecting 85% of Pugs. A moderate correlation (r = 0.50) was found between duration of clinical signs and outcome. In 8 dogs with deteriorating clinical signs, follow-up MRI revealed regrowth of the SAD in 2 cases, new SAD formation in 2 cases, and intramedullary T2W hyperintensity/syringomyelia in 6 cases. CONCLUSIONS AND CLINICAL IMPORTANCE: This study suggests that Pugs with thoracolumbar SAD do not have a favorable long-term prognosis after surgical treatment for reasons yet to be determined.


Subject(s)
Arachnoid Cysts/veterinary , Dog Diseases/surgery , Animals , Arachnoid Cysts/diagnostic imaging , Arachnoid Cysts/surgery , Dogs , Female , Magnetic Resonance Imaging/veterinary , Male , Retrospective Studies , Syringomyelia/veterinary , Treatment Outcome
15.
PLoS One ; 13(9): e0204073, 2018.
Article in English | MEDLINE | ID: mdl-30235266

ABSTRACT

Ciliopathies presenting as inherited hepatorenal fibrocystic disorders are rare in humans and in dogs. We describe here a novel lethal ciliopathy in Norwich Terrier puppies that was diagnosed at necropsy and characterized as diffuse cystic renal disease and hepatic fibrosis. The histopathological findings were typical for cystic renal dysplasia in which the cysts were located in the straight portion of the proximal tubule, and thin descending and ascending limbs of Henle's loop. The pedigree of the affected puppies was suggestive of an autosomal recessive inheritance and therefore, whole exome sequencing and homozygosity mapping were used for identification of the causative variant. The analyses revealed a case-specific homozygous splice donor site variant in a cilia related gene, INPP5E: c.1572+5G>A. Association of the variant with the defect was validated in a large cohort of Norwich Terriers with 3 cases and 480 controls, the carrier frequency being 6%. We observed that the identified variant introduces a novel splice site in INPP5E causing a frameshift and formation of a premature stop codon. In conclusion, our results suggest that the INPP5E: c.1572+5G>A variant is causal for the ciliopathy in Norwich Terriers. Therefore, genetic testing can be carried out in the future for the eradication of the disease from the breed.


Subject(s)
Liver Cirrhosis/enzymology , Liver Cirrhosis/genetics , Mutation/genetics , Phosphoric Monoester Hydrolases/genetics , Polycystic Kidney, Autosomal Recessive/enzymology , Polycystic Kidney, Autosomal Recessive/genetics , RNA Splice Sites/genetics , Animals , Cilia/metabolism , Dogs , Epithelial Cells/metabolism , Female , Gene Expression Regulation , Homozygote , Kidney/pathology , Liver Cirrhosis/pathology , Male , Organogenesis , Pedigree , Phosphoric Monoester Hydrolases/metabolism , Polycystic Kidney, Autosomal Recessive/pathology , RNA, Messenger/genetics , RNA, Messenger/metabolism , Exome Sequencing
16.
Mol Ther ; 26(9): 2243-2254, 2018 09 05.
Article in English | MEDLINE | ID: mdl-30017877

ABSTRACT

Lymphodepleting preconditioning with high-dose chemotherapy is commonly used to increase the clinical efficacy of adoptive T cell therapy (ACT) strategies, however, with severe toxicity for patients. Conversely, oncolytic adenoviruses are safe and, when engineered to express interleukin-2 (IL-2) and tumor necrosis factor alpha (TNF-α), they can achieve antitumor immunomodulatory effects similar to lymphodepletion. Therefore, we compare the safety and efficacy of such adenoviruses with a cyclophosphamide- and fludarabine-containing lymphodepleting regimen in the setting of ACT. Human adenovirus (Ad5/3-E2F-D24-hTNF-α-IRES-hIL-2; TILT-123) replication was studied using a Syrian hamster pancreatic tumor model (HapT1) infused with tumor-infiltrating lymphocytes (TILs). Using the oncolytic virus instead of lymphodepletion resulted in superior efficacy and survival. Immune cells responsive to TNF-α IL-2 were studied using an immunocompetent mouse melanoma model (B16.OVA) infused with ovalbumin-specific T (OT-I) cells. Here, the adenovirus approach improved tumor control together with increased intratumoral Th1 cytokine levels and infiltration of CD8+ T cells and CD86+ dendritic cells. Similar to humans, lymphodepleting preconditioning caused severe cytopenias, systemic inflammation, and damage to vital organs. Toxicity was minimal in adenovirus- and OT-I-treated mice. These findings demonstrate that ACT can be effectively facilitated by cytokine-coding adenovirus without requiring lymphodepletion, a rationale being clinically investigated.


Subject(s)
Interleukin-2/therapeutic use , Tumor Necrosis Factor-alpha/therapeutic use , Adenoviridae/genetics , Animals , Cell Line , Disease Models, Animal , Genetic Vectors/genetics , Humans , Immunotherapy, Adoptive/methods , Male , Melanoma/immunology , Melanoma/therapy , Mesocricetus , Mice , T-Lymphocytes/metabolism , T-Lymphocytes/physiology
17.
Int J Cancer ; 141(7): 1458-1468, 2017 10 01.
Article in English | MEDLINE | ID: mdl-28614908

ABSTRACT

Systemic high dose interleukin-2 (IL-2) postconditioning has long been utilized in boosting the efficacy of T cells in adoptive cell therapy (ACT) of solid tumors. The resulting severe off-target toxicity of these regimens renders local production at the tumor an attractive concept with possible safety gains. We evaluated the efficacy and safety of intratumorally administered IL-2-coding adenoviruses in combination with tumor-infiltrating lymphocyte therapy in syngeneic Syrian hamsters bearing HapT1 pancreatic tumors and with T cell receptor transgenic ACT in B16.OVA melanoma bearing C57BL/6 mice. The models are complementary: hamsters are semi-permissive for human oncolytic adenovirus, whereas detailed immunological analyses are possible in mice. In both models, local production of IL-2 successfully replaced the need for systemic recombinant IL-2 (rIL-2) administration and increased the efficacy of the cell therapy. Furthermore, vectored delivery of IL-2 significantly enhanced the infiltration of CD8+ T cells, M1-like macrophages, and B-cells while systemic rIL-2 increased CD25 + FoxP3+ T cells at the tumor. In contrast with vectored delivery, histopathological analysis of systemic rIL-2-treated animals revealed significant changes in lungs, livers, hearts, spleens, and kidneys. In summary, local IL-2 production results in efficacy and safety gains in the context of ACT. These preclinical assessments provide the rationale for ongoing clinical translation.


Subject(s)
Adenoviridae/metabolism , Immunotherapy, Adoptive/methods , Interleukin-2/biosynthesis , Lymphocytes, Tumor-Infiltrating/immunology , Melanoma, Experimental/therapy , Pancreatic Neoplasms/therapy , Adenoviridae/immunology , Adoptive Transfer/methods , Animals , B-Lymphocytes/immunology , CD8-Positive T-Lymphocytes/immunology , Cell Movement/immunology , Cricetinae , Disease Models, Animal , Female , Forkhead Transcription Factors/immunology , Genetic Vectors , Inflammation Mediators/blood , Interleukin-2/administration & dosage , Interleukin-2/immunology , Interleukin-2 Receptor alpha Subunit/immunology , Lung/blood supply , Lung/pathology , Lymphocytes, Tumor-Infiltrating/transplantation , Macrophages/immunology , Male , Melanoma, Experimental/immunology , Mesocricetus , Mice , Mice, Inbred C57BL , Pancreatic Neoplasms/immunology , Random Allocation , Recombinant Proteins/administration & dosage , Recombinant Proteins/immunology
18.
PLoS One ; 12(5): e0177527, 2017.
Article in English | MEDLINE | ID: mdl-28493971

ABSTRACT

A rare hereditary mechanobullous disorder called epidermolysis bullosa (EB) causes blistering in the skin and the mucosal membranes. To date, nineteen EB-related genes have been discovered in human and other species. We describe here a novel EB variant in dogs. Two newborn littermates of Central Asian Shepherd dogs with severe signs of skin blistering were brought to a veterinary clinic and euthanized due to poor prognosis. In post-mortem examination, the puppies were shown to have findings in the skin and the mucosal membranes characteristic of EB. A whole-genome sequencing of one of the affected puppies was performed to identify the genetic cause. The resequencing data were filtered under a recessive model against variants from 31 other dog genomes, revealing a homozygous case-specific nonsense variant in one of the known EB-causing genes, COL7A1 (c.4579C>T, p.R1527*). The variant results in a premature stop codon and likely absence of the functional protein in the basement membrane of the skin in the affected dogs. This was confirmed by immunohistochemistry using a COL7A1 antibody. Additional screening of the variant indicated full penetrance and breed specificity at ~28% carrier frequency. In summary, this study reveals a novel COL7A1 variant causing recessive dystrophic EB and provides a genetic test for the eradication of the disease from the breed.


Subject(s)
Codon, Nonsense/genetics , Collagen Type VII/genetics , Epidermolysis Bullosa Dystrophica/genetics , Epidermolysis Bullosa Dystrophica/veterinary , Animals , Asia , Base Sequence , Case-Control Studies , Cohort Studies , Dogs , Epidermolysis Bullosa Dystrophica/pathology , Female , Genome , Immunohistochemistry , Male , Pedigree
19.
BMC Vet Res ; 11: 170, 2015 Jul 28.
Article in English | MEDLINE | ID: mdl-26215394

ABSTRACT

BACKGROUND: Dogs suffer from spontaneous tumors which may be amenable to therapies developed for human cancer patients, and dogs may serve as large-animal cancer models. A non-pathogenic Semliki Forest virus vector VA7-EGFP previously showed promise in targeting human tumor xenografts in mice, but the oncolytic capacity of the virus in canine cancer cells and the safety of the virus in higher mammals such as dogs, are not known. We therefore assessed the oncolytic potency of VA7-EGFP against canine cancer cells by infectivity and viability assays in two dog solid tumor cell lines. Furthermore we performed a 3-week safety study in two adult Beagles which received a single intravenous injection of ~2 × 10(5) plaque forming units of parental A7(74) strain. RESULTS: VA7-EGFP was able to replicate in and kill both canine cancer cell lines tested. No adverse events were observed in either of the two virus-injected adult Beagles and no infective virus could be recovered from any of the biological samples collected over the course of the study. Neutralizing antibodies to Semliki Forest virus became detectable in the dogs at 5 days post infection and remained elevated until study termination. CONCLUSIONS: Based on these results, testing of the oncolytic potential of attenuated Semliki Forest virus in canine cancer patients appears feasible.


Subject(s)
Cancer Vaccines/adverse effects , Dog Diseases/chemically induced , Animals , Antibodies, Neutralizing/blood , Cell Line, Tumor , Dogs , Female , Semliki forest virus , Virus Replication/physiology
20.
J Clin Microbiol ; 53(9): 3047-53, 2015 Sep.
Article in English | MEDLINE | ID: mdl-26085616

ABSTRACT

We present an unusual equine endometritis case associated with Cladophialophora bantiana in a 15-year-old mare. The mare displayed infertility and uterine fluid accumulation with numerous black, hairy granules. Microscopically, the fluid revealed numerous septate, dark fungal hyphae and conidia in chains. Culture yielded C. bantiana (CBS 138271); the species was confirmed by internal transcribed spacer (ITS) sequencing. Treatment was unsuccessful. C. bantiana causes cerebral phaeohyphomycosis in humans, while animal cases are rare. Animal cases are reviewed.


Subject(s)
Ascomycota/isolation & purification , Endometritis/pathology , Endometritis/veterinary , Horse Diseases/microbiology , Horse Diseases/pathology , Mycoses/pathology , Mycoses/veterinary , Animals , Ascomycota/classification , Ascomycota/genetics , Cluster Analysis , DNA, Fungal/chemistry , DNA, Fungal/genetics , DNA, Ribosomal Spacer/chemistry , DNA, Ribosomal Spacer/genetics , Endometritis/microbiology , Exudates and Transudates/microbiology , Female , Horses , Microbiological Techniques , Microscopy , Molecular Sequence Data , Mycoses/microbiology , Phylogeny , Pregnancy , Sequence Analysis, DNA
SELECTION OF CITATIONS
SEARCH DETAIL
...