Network


Latest external collaboration on country level. Dive into details by clicking on the dots.

Hotspot


Dive into the research topics where Albert Stanek is active.

Publication


Featured researches published by Albert Stanek.


American Journal of Obstetrics and Gynecology | 1984

The relationship between human papillomavirus and lower genital intraepithelial neoplasia in immunosuppressed women

Sillman Fh; Albert Stanek; Alexander Sedlis; Julian Rosenthal; Karl W. Lanks; Dorothy Buchhagen; Anthony D. Nicastri; John Boyce

In a group of 20 immunosuppressed women with lower genital neoplasia, evidence of associated human papillomaviral infection was found in all patients on the basis of the histologic identification of koilocytes in the upper strata of areas of mild or moderate dysplasia. Immunohistochemical study of similar areas disclosed human papilloma structural antigens in the lesions in 60%, while 50% had lesions in which human papilloma virions were detected by the electron microscope. An abnormal immunologic status, indicated by an altered T-helper/T-suppressor ratio, a deficient response to mitogenic stimulation, or both, was confirmed in 80% of the patients studied. Twelve of the 20 patients had unusually persistent and recurrent intraepithelial neoplasia, and in one the disorder progressed to invasive epidermoid carcinoma. The progressive behavior of human papillomavirus-associated neoplasia in these immunosuppressed patients might represent an accelerated version of the long-term course of such lesions in immunocompetent hosts.


Gastrointestinal Endoscopy | 1985

Pancreatic tuberculosis mimicking carcinoma

Kota L. Chandrasekhara; Swaminath K. Iyer; Albert Stanek; Horace Herbstman

5. Eliopoulos GM, Moellering RC. Differentiation of bacterial endocarditis caused by Streptococcus bouis: clinical significance. Intern Med Specialist 1982;3:63-6. 6. Brooks RJ, Ravreby WD, Keusch G, Bottone E. More on Streptococcus bouis endocarditis and bowel carcinoma. N Engl J Med 1978;298:572. 7. Herrington P, Finkelman D, Balart L, Hines C, Ferrante W. Streptococcus bouis septicemia and pancreatic adenocarcinoma. Ann Intern Med 1980;92:441. 8. Klein RS, Recco RA, Catalano MT, Edberg SC, Casey Jr,


World Journal of Gastrointestinal Pathophysiology | 2016

Mechanisms of interleukin-22's beneficial effects in acute pancreatitis.

Chongmin Huan; Daniel Kim; Peiqi Ou; Antonio E. Alfonso; Albert Stanek

Acute pancreatitis (AP) is a disorder characterized by parenchymal injury of the pancreas controlled by immune cell-mediated inflammation. AP remains a significant challenge in the clinic due to a lack of specific and effective treatment. Knowledge of the complex mechanisms that regulate the inflammatory response in AP is needed for the development of new approaches to treatment, since immune cell-derived inflammatory cytokines have been recognized to play critical roles in the pathogenesis of the disease. Recent studies have shown that interleukin (IL)-22, a cytokine secreted by leukocytes, when applied in the severe animal models of AP, protects against the inflammation-mediated acinar injury. In contrast, in a mild AP model, endogenous IL-22 has been found to be a predominantly anti-inflammatory mediator that inhibits inflammatory cell infiltration via the induction of Reg3 proteins in acinar cells, but does not protect against acinar injury in the early stage of AP. However, constitutively over-expressed IL-22 can prevent the initial acinar injury caused by excessive autophagy through the induction of the anti-autophagic proteins Bcl-2 and Bcl-XL. Thus IL-22 plays different roles in AP depending on the severity of the AP model. This review focuses on these recently reported findings for the purpose of better understanding IL-22s regulatory roles in AP which could help to develop a novel therapeutic strategy.


Pancreas | 2008

Small-interference RNA gene knockdown of pancreatitis-associated proteins in rat acute pancreatitis.

Yin Yao Lin; Domenico Viterbo; Cathy M. Mueller; Albert Stanek; Tamar A. Smith-Norowitz; Hazel Drew; Raj Wadgaonkar; Michael E. Zenilman; Martin H. Bluth

Objectives: Pancreatitis-associated proteins (PAPs) are induced in acute pancreatitis and antisense-mediated gene knockdown of PAP decreased PAP gene expression and worsened pancreatitis. Here, we investigated the effect of a more stable inhibition of PAP using small-interference RNA gene knockdown in vitro and in an in vivo model of experimental pancreatitis. Methods: Pancreatitis-associated protein-specific siRNA was administered to AR42J cell cultures or rats induced with pancreatitis. Controls included administration of scrambled siRNA or vehicle alone. After 24 hours, cells and pancreata were harvested and assessed for PAP (PAP 1, PAP 2, PAP 3) gene expression and pancreatitis severity. Results: In vitro, PAP protein, and mRNA levels were reduced (PAP 1, 76%; PAP 2, 8%; PAP 3, 24%) in cells treated with PAP siRNA. In vivo, PAP 1, and PAP 3 expressions were reduced (PAP 1, 36%; PAP 3, 66%) in siRNA-treated rats; there was no difference in PAP 2 isoform mRNA expression and serum protein levels. Serum amylase and lipase levels decreased (≥50%) after administration of siRNA; interleukin (IL) 1&bgr;, IL-4, and IL-6 increased, whereas C-reactive protein and tumor necrosis factor-&agr; decreased when compared with vehicle control. Administration of PAP siRNA correlated with worsening histopathology. Conclusions: siRNA-mediated gene knockdown of PAP worsens pancreatitis. Differences in gene knockdown technology may provide different approaches to study gene function.


The American Journal of Medicine | 1986

Spontaneous rupture of the stomach

Kota L. Chandrasekhara; Swaminath K. Iyer; Alfred L. Sutton; Albert Stanek

A case of spontaneous rupture of the stomach associated with perforation of the diaphragm is reported. Spontaneous rupture of the stomach is a rare but fatal condition. The pathophysiologic features, clinical manifestations, and treatment are discussed.


World Journal of Gastroenterology | 2012

Acute pancreatitis in aging animals: Loss of pancreatitis-associated protein protection?

Sophia L. Fu; Albert Stanek; Cathy M. Mueller; Nefertti A Brown; Chongmin Huan; Martin H. Bluth; Michael E. Zenilman

AIM To investigate the effect of age on severity of acute pancreatitis (AP) using biochemical markers, histology and expression of the protective pancreatitis-associated proteins (PAPs). METHODS AP was induced via intraductal injection of 4% sodium taurocholate in young and old rats. Sera and pancreata were assayed at 24 h for the parameters listed above; we also employed a novel molecular technique to assess bacterial infiltration using polymerase chain reaction to measure bacterial genomic ribosomal RNA. RESULTS At 24 h after induction of AP, the pancreata of older animals had less edema (mean ± SE histologic score of young vs old: 3.11 ± 0.16 vs 2.50 ± -0.11, P < 0.05), decreased local inflammatory response (histologic score of stromal infiltrate: 3.11 ± 0.27 vs 2.00 ± 0.17, P < 0.05) and increased bacterial infiltration (174% ± 52% increase from sham vs 377% ± 4%, P < 0.05). A decreased expression of PAP1 and PAP2 was demonstrated by Western blotting analysis and immunohistochemical staining. There were no differences in serum amylase and lipase activity, or tissue myeloperoxidase or monocyte chemotactic protein-1 levels. However, in the most-aged group, serum C-reactive protein levels were higher (young vs old: 0.249 ± 0.04 mg/dL vs 2.45 ± 0.68 mg/dL, P < 0.05). CONCLUSION In older animals, there is depressed PAP expression related to a blunted inflammatory response in AP which is associated with worsened bacterial infiltration and higher C-reactive protein level; this may explain the more aggressive clinical course.


Hepatology | 2016

Liver serine palmitoyltransferase activity deficiency in early life impairs adherens junctions and promotes tumorigenesis

Zhiqiang Li; Inamul Kabir; Hui Jiang; Hongwen Zhou; Jenny Libien; Jianying Zeng; Albert Stanek; Peiqi Ou; Kailyn R. Li; Shane Zhang; Hai H. Bui; Ming-Shang Kuo; Tae-Sik Park; Benjamin Kim; Tilla S. Worgall; Chongmin Huan; Xian-Cheng Jiang

Serine palmitoyltransferase is the key enzyme in sphingolipid biosynthesis. Mice lacking serine palmitoyltransferase are embryonic lethal. We prepared liver‐specific mice deficient in the serine palmitoyltransferase long chain base subunit 2 gene using an albumin‐cyclization recombination approach and found that the deficient mice have severe jaundice. Moreover, the deficiency impairs hepatocyte polarity, attenuates liver regeneration after hepatectomy, and promotes tumorigenesis. Importantly, we show that the deficiency significantly reduces sphingomyelin but not other sphingolipids in hepatocyte plasma membrane; greatly reduces cadherin, the major protein in adherens junctions, on the membrane; and greatly induces cadherin phosphorylation, an indication of its degradation. The deficiency affects cellular distribution of β‐catenin, the central component of the canonical Wnt pathway. Furthermore, such a defect can be partially corrected by sphingomyelin supplementation in vivo and in vitro. Conclusion: The plasma membrane sphingomyelin level is one of the key factors in regulating hepatocyte polarity and tumorigenesis. (Hepatology 2016;64:2089‐2102).


World Journal of Gastrointestinal Pathophysiology | 2018

Acinar cell injury induced by inadequate unfolded protein response in acute pancreatitis

Kaylene Barrera; Albert Stanek; Kei Okochi; Zuzanna Niewiadomska; Cathy M. Mueller; Peiqi Ou; Devon John; Antonio E. Alfonso; Scott Tenner; Chongmin Huan

Acute pancreatitis (AP) is an inflammatory disorder of pancreatic tissue initiated in injured acinar cells. Severe AP remains a significant challenge due to the lack of effective treatment. The widely-accepted autodigestion theory of AP is now facing challenges, since inhibiting protease activation has negligible effectiveness for AP treatment despite numerous efforts. Furthermore, accumulating evidence supports a new concept that malfunction of a self-protective mechanism, the unfolded protein response (UPR), is the driving force behind the pathogenesis of AP. The UPR is induced by endoplasmic reticulum (ER) stress, a disturbance frequently found in acinar cells, to prevent the aggravation of ER stress that can otherwise lead to cell injury. In addition, the UPR’s signaling pathways control NFκB activation and autophagy flux, and these dysregulations cause acinar cell inflammatory injury in AP, but with poorly understood mechanisms. We therefore summarize the protective role of the UPR in AP, propose mechanistic models of how inadequate UPR could promote NFκB’s pro-inflammatory activity and impair autophagy’s protective function in acinar cells, and discuss its relevance to current AP treatment. We hope that insight provided in this review will help facilitate the research and management of AP.


Cancer Research | 2012

Abstract B25: Drug-eluting microparticles for the treatment of pancreatic cancer: Preliminary in vivo results.

Amon Asgharpour; Manoj Ganesh; Jing Ling; Albert Stanek; Wenchun Xie; Alicia Gooding; Sherif A. Andrawes; Richard A. Gross; Frank G. Gress; Laura Martello-Rooney

Pancreatic cancer is the fourth leading cause of cancer death in the U.S. Current treatment regimens have had a minimal impact on altering the course of the disease, establishing the need for alternative modalities of therapy. One of the main issues with systemic chemotherapy is in vivo data demonstrating compromised blood flow to the pancreatic tumor offering one explanation for the lack of efficacy in pancreatic cancer patients. A novel option would be to deliver drug directly to the pancreatic tumor over a sustained period of time to circumvent this barrier, and at the same time decrease the side effects associated with systemic delivery. Here we explore the feasibility of direct injection of biodegradable polymer-based microparticles (MPs) with an approximate size of 10 μm into the tail portion of the mouse pancreas. A laparotomy was performed on C57BL/6 mice to expose the pancreas followed by injection of 50 μl phosphate-buffered saline (PBS), 50 μl blank MPs/PBS or 25 μl blank MPs/PBS into the pancreatic tail using a 29-gauge needle (165 μm inner diameter). The mice were sacrificed at the following post-operative timepoints: 24 hrs, 3 days and 7 days. The mice were weighed daily and blood was drawn pre- and post-operatively for pancreatic enzyme testing. Mouse tissue samples of the pancreas, liver, spleen and duodenum were placed in formalin upon sacrifice. All of the mice survived the surgery and exhibited minimal weight loss, which was reversed by day 7. Lipase and amylase levels were mildly elevated after 24 hrs, but returned to pre-bleed levels by day 3. The analysis of the pancreatic tissue sections disclosed acinar cell damage only in the area surrounding the injection site and MP deposit. There was no evidence of pancreatitis, a concern following manipulation of the pancreas. No indication of MP migration was observed in sections of the liver, spleen or duodenum. These findings established that direct injection of MPs into the mouse pancreas was feasible and safe and supported proceeding to the next phase of utilizing drug-loaded MPs in a mouse model of pancreatic cancer. An orthotopic nude mouse model of pancreatic cancer was employed by injecting PANC-1 human pancreatic cancer cells into the tail section of the mouse pancreas via laparotomy. Two weeks post-cancer cell injection, a second laparotomy was performed to inject 50 μl drug-loaded MPs (average size range 25-50 μm) or 50 μl PBS into the same tail section of the pancreas. The mice were weighed 3 times weekly and blood was drawn pre- and postoperatively to measure pancreatic enzyme levels. For HPLC detection of drug concentration, mouse plasma and tissue samples were collected from control and treated mice at 4 weeks post-MP injection. Mouse tissue samples also were taken to evaluate the local effects of constant drug release on the pancreatic tumors and to determine the extent of drug delivery to the spleen and liver. Positive results of these combined studies will justify additional preclinical investigation in a transgenic mouse model of pancreatic cancer, with the final objective to validate the potential use of drug-eluting MPs to deliver localized tumor treatment for patients with pancreatic cancer. Citation Format: Amon Asgharpour, Manoj Ganesh, Jing Ling, Albert Stanek, Wenchun Xie, Alicia Gooding, Sherif Andrawes, Richard Gross, Frank Gress, Laura Martello-Rooney. Drug-eluting microparticles for the treatment of pancreatic cancer: Preliminary in vivo results. [abstract]. In: Proceedings of the AACR Special Conference on Pancreatic Cancer: Progress and Challenges; Jun 18-21, 2012; Lake Tahoe, NV. Philadelphia (PA): AACR; Cancer Res 2012;72(12 Suppl):Abstract nr B25.


Gastroenterology | 2009

635 Acute Pancreatitis (AP) in Aging Animals-Loss of PAP Protection?

Sophia L. Fu; Albert Stanek; E. Hassanain; Okiremute Oyiborhoro; Cathy M. Mueller; Nefertiti A. Brown; Chongmin Huan; Michael E. Zenilman

Background: Although acute inflammation is a critical protective response against injury and infection, persistent inflammation and the resulting oxidative stress drive the pathogenesis of numerous chronic degenerative diseases, including carcinogenesis. In human pancreatic ductal adenocarcinoma (PDA) and in animal models that recapitulate the disease progression, an intense fibroinflammatory reaction composed of stromal and immune cells accompanies the progression from normal histology to PDA. Angiotensin II (AngII), the principal hormone of the renin angiotensin system, is actively generated in the pancreas and has been proposed as a key mediator of inflammation. Monocyte chemoattractant protein(MCP)-1 is a chemokine that plays an important role in the recruitment of mononuclear cells into sites of inflammation. Objective: To investigate the potential proinflammatory role of AngII in PDA through studying its effect on MCP-1. Methods: PDA cells (AsPC-1, HS766T, MiaPaca) were cultured and treated with or without Ang II (10-8-10-6mol/L), in the presence or absence of AngII type 1 receptor blocker, losartan, or AngII type 2 receptor blocker, PD123319. MCP-1 mRNA was analyzed by real time PCR and its protein by ELISA. Luciferase-labeled promoter studies evaluated the effect of AngII on the transcription of MCP-1 and nuclear factor-κB (NF-κB). The effect of receptor blockers on the endogenous and AngII-induced activation and nuclear translocation of NF-kB was examined by luminescence assay and immunohistochemistry. Results: AngII significantly increased the expression ofMCP-1mRNA and protein in PDA cells, and induced its promoter activity. Constitutive and AngII-induced MCP-1 mRNA and promoter activity were significantly reduced in the presence of losartan but were unchanged by PD123319. AngII induced the activation and nuclear translocation of NF-kB, an effect that was inhibited by losartan, but not PD 1233319. Blocking NF-kB activation by pyrrolidine dithiocarbamate decreased the AngII-mediated increase in MCP1 mRNA. Conclusions: Our data provide a novel insight into an AngII-initiated signal transduction pathway that regulates MCP-1 as a possible inflammatory mechanism in PDA, and suggest that AngII blockade may regulate chemokine-induced signal transduction to prevent or reduce inflammation in PDA.

Collaboration


Dive into the Albert Stanek's collaboration.

Top Co-Authors

Avatar

Cathy M. Mueller

SUNY Downstate Medical Center

View shared research outputs
Top Co-Authors

Avatar

Chongmin Huan

SUNY Downstate Medical Center

View shared research outputs
Top Co-Authors

Avatar

Antonio E. Alfonso

SUNY Downstate Medical Center

View shared research outputs
Top Co-Authors

Avatar

Michael E. Zenilman

SUNY Downstate Medical Center

View shared research outputs
Top Co-Authors

Avatar

Peiqi Ou

SUNY Downstate Medical Center

View shared research outputs
Top Co-Authors

Avatar

Martin H. Bluth

SUNY Downstate Medical Center

View shared research outputs
Top Co-Authors

Avatar

Alexander Sedlis

SUNY Downstate Medical Center

View shared research outputs
Top Co-Authors

Avatar

John Boyce

SUNY Downstate Medical Center

View shared research outputs
Top Co-Authors

Avatar

M.H. Bluth

SUNY Downstate Medical Center

View shared research outputs
Top Co-Authors

Avatar

Richard A. Gross

Rensselaer Polytechnic Institute

View shared research outputs
Researchain Logo
Decentralizing Knowledge