41 results on '"Dinesh Jaishankar"'
Search Results
2. Archaic connectivity between the sulfated heparan sulfate and the herpesviruses – An evolutionary potential for cross-species interactions
- Author
-
James Elste, Angelica Chan, Chandrashekhar Patil, Vinisha Tripathi, Daniel M. Shadrack, Dinesh Jaishankar, Andrew Hawkey, Michelle Swanson Mungerson, Deepak Shukla, and Vaibhav Tiwari
- Subjects
Heparan sulfare ,Virus entry ,Virus cell-to-cell spread ,Virus cell fusion ,Sulphated heparan sulfate ,Biotechnology ,TP248.13-248.65 - Abstract
The structural diversity of metazoic heparan sulfate (HS) composed of unique sulfated domains is remarkably preserved among various vertebrates and invertebrate species. Interestingly the sulfated moieties of HS have been known as the key determinants generating extraordinary ligand binding sites in the HS chain to regulate multiple biological functions and homeostasis. One such ligand for 3-O sulfation in the HS chain is a glycoprotein D (gD) from an ancient herpesvirus, herpes simplex virus (HSV). This interaction between gD and 3-O sulfated HS leads to virus-cell fusion to promote HSV entry. It is quite astonishing that HSV-1, which infects two-thirds of the world population, is also capable of causing severe diseases in primates and non-primates including primitive zebrafish. Supporting evidence that HSV may cross the species barrier comes from the fact that an enzymatic modification in HS encoded by 3-O sulfotransferase-3 (3-OST-3) from a vertebrate zoonotic species enhances HSV-1 infectivity. The latter phenomenon suggests the possible role of sulfated-HS as an entry receptor during reverse zoonosis, especially during an event when humans encounter domesticated animals in proximity. In this mini-review, we explore the possibility that structural diversity in HS may have played a substantial role in species-specific adaptability for herpesviruses in general including their potential role in promoting cross-species transmission.
- Published
- 2023
- Full Text
- View/download PDF
3. Skin Infiltrate Composition as a Telling Measure of Responses to Checkpoint Inhibitors
- Author
-
Cory Kosche, Dinesh Jaishankar, Cormac Cosgrove, Prathyaya Ramesh, Suyeon Hong, Lin Li, Rohan S. Shivde, Deven Bhuva, Bethany E. Perez White, Sabah S. Munir, Hui Zhang, Kurt Q. Lu, Jennifer N. Choi, and I. Caroline Le Poole
- Subjects
Dermatology ,RL1-803 - Abstract
Checkpoint inhibitors treat a variety of tumor types with significant benefits. Unfortunately, these therapies come with diverse adverse events. Skin rash is observed early into treatment and might serve as an indicator of downstream responses to therapy. We studied the cellular composition of cutaneous eruptions and whether their contribution varies with the treatment applied. Skin samples from 18 patients with cancer and 11 controls were evaluated by mono- and multiplex imaging, quantification, and statistical analysis. T cells were the prime contributors to skin rash, with T cells and macrophages interacting and proliferating on site. Among T cell subsets examined, type 1 and 17 T cells were relatively increased among inflammatory skin infiltrates. A combination of increased cytotoxic T cell content and decreased macrophage abundance was associated with dual checkpoint inhibition over PD1 inhibition alone. Importantly, responders significantly separated from nonresponders by greater CD68+ macrophage and either CD11c+ antigen-presenting cell or CD4+ T cell abundance in skin rash. The microenvironment promoted epidermal proliferation and thickening as well. The combination of checkpoint inhibitors used affects the development and composition of skin infiltrates, whereas the combined abundance of two cell types in cutaneous eruptions aligns with responses to checkpoint inhibitor therapy.
- Published
- 2023
- Full Text
- View/download PDF
4. Editorial: Ocular infection of herpes: Immunology, pathogenesis, and interventions
- Author
-
Tejabhiram Yadavalli, Chandrashekhar Patil, Dinesh Jaishankar, and Rahul Suryawanshi
- Subjects
herpes ,pathogenesis ,vaccines ,eye ,cornea ,trigeminal ganglia ,Microbiology ,QR1-502 - Published
- 2022
- Full Text
- View/download PDF
5. Benign tumors in TSC are amenable to treatment by GD3 CAR T cells in mice
- Author
-
Ancy Thomas, Saurav Sumughan, Emilia R. Dellacecca, Rohan S. Shivde, Nicola Lancki, Zhussipbek Mukhatayev, Cristina C. Vaca, Fei Han, Levi Barse, Steven W. Henning, Jesus Zamora-Pineda, Suhail Akhtar, Nikhilesh Gupta, Jasmine O. Zahid, Stephanie R. Zack, Prathyaya Ramesh, Dinesh Jaishankar, Agnes S.Y. Lo, Joel Moss, Maria M. Picken, Thomas N. Darling, Denise M. Scholtens, Daniel F. Dilling, Richard P. Junghans, and I. Caroline Le Poole
- Subjects
Immunology ,Therapeutics ,Medicine - Abstract
Mutations underlying disease in tuberous sclerosis complex (TSC) give rise to tumors with biallelic mutations in TSC1 or TSC2 and hyperactive mammalian target of rapamycin complex 1 (mTORC1). Benign tumors might exhibit de novo expression of immunogens, targetable by immunotherapy. As tumors may rely on ganglioside D3 (GD3) expression for mTORC1 activation and growth, we compared GD3 expression in tissues from patients with TSC and controls. GD3 was overexpressed in affected tissues from patients with TSC and also in aging Tsc2+/– mice. As GD3 overexpression was not accompanied by marked natural immune responses to the target molecule, we performed preclinical studies with GD3 chimeric antigen receptor (CAR) T cells. Polyfunctional CAR T cells were cytotoxic toward GD3-overexpressing targets. In mice challenged with Tsc2–/– tumor cells, CAR T cells substantially and durably reduced the tumor burden, correlating with increased T cell infiltration. We also treated aged Tsc2+/– heterozygous (>60 weeks) mice that carry spontaneous Tsc2–/– tumors with GD3 CAR or untransduced T cells and evaluated them at endpoint. Following CAR T cell treatment, the majority of mice were tumor free while all control animals carried tumors. The outcomes demonstrate a strong treatment effect and suggest that targeting GD3 can be successful in TSC.
- Published
- 2021
- Full Text
- View/download PDF
6. Disruption of innate defense responses by endoglycosidase HPSE promotes cell survival
- Author
-
Alex Agelidis, Benjamin A. Turturice, Rahul K. Suryawanshi, Tejabhiram Yadavalli, Dinesh Jaishankar, Joshua Ames, James Hopkins, Lulia Koujah, Chandrashekhar D. Patil, Satvik R. Hadigal, Evan J. Kyzar, Anaamika Campeau, Jacob M. Wozniak, David J. Gonzalez, Israel Vlodavsky, Jin-ping Li, David L. Perkins, Patricia W. Finn, and Deepak Shukla
- Subjects
Cell biology ,Microbiology ,Medicine - Abstract
The drive to withstand environmental stresses and defend against invasion is a universal trait extant in all forms of life. While numerous canonical signaling cascades have been characterized in detail, it remains unclear how these pathways interface to generate coordinated responses to diverse stimuli. To dissect these connections, we followed heparanase (HPSE), a protein best known for its endoglycosidic activity at the extracellular matrix but recently recognized to drive various forms of late-stage disease through unknown mechanisms. Using herpes simplex virus-1 (HSV-1) infection as a model cellular perturbation, we demonstrate that HPSE acts beyond its established enzymatic role to restrict multiple forms of cell-intrinsic defense and facilitate host cell reprogramming by the invading pathogen. We reveal that cells devoid of HPSE are innately resistant to infection and counteract viral takeover through multiple amplified defense mechanisms. With a unique grasp of the fundamental processes of transcriptional regulation and cell death, HPSE represents a potent cellular intersection with broad therapeutic potential.
- Published
- 2021
- Full Text
- View/download PDF
7. Antigen Specificity Enhances Disease Control by Tregs in Vitiligo
- Author
-
Zhussipbek Mukhatayev, Emilia R. Dellacecca, Cormac Cosgrove, Rohan Shivde, Dinesh Jaishankar, Katherine Pontarolo-Maag, Jonathan M. Eby, Steven W. Henning, Yekaterina O. Ostapchuk, Kettil Cedercreutz, Alpamys Issanov, Shikhar Mehrotra, Andreas Overbeck, Richard P. Junghans, Joseph R. Leventhal, and I. Caroline Le Poole
- Subjects
vitiligo ,regulatory T cells ,chimeric antigen receptor T cells ,ganglioside D3 ,antigen-specific Treg ,autoimmune diseases ,Immunologic diseases. Allergy ,RC581-607 - Abstract
Vitiligo is an autoimmune skin disease characterized by melanocyte destruction. Regulatory T cells (Tregs) are greatly reduced in vitiligo skin, and replenishing peripheral skin Tregs can provide protection against depigmentation. Ganglioside D3 (GD3) is overexpressed by perilesional epidermal cells, including melanocytes, which prompted us to generate GD3-reactive chimeric antigen receptor (CAR) Tregs to treat vitiligo. Mice received either untransduced Tregs or GD3-specific Tregs to test the hypothesis that antigen specificity contributes to reduced autoimmune reactivity in vitro and in vivo. CAR Tregs displayed increased IL-10 secretion in response to antigen, provided superior control of cytotoxicity towards melanocytes, and supported a significant delay in depigmentation compared to untransduced Tregs and vehicle control recipients in a TCR transgenic mouse model of spontaneous vitiligo. The latter findings were associated with a greater abundance of Tregs and melanocytes in treated mice versus both control groups. Our data support the concept that antigen-specific Tregs can be prepared, used, and stored for long-term control of progressive depigmentation.
- Published
- 2020
- Full Text
- View/download PDF
8. The relationship between stress and vitiligo: Evaluating perceived stress and electronic medical record data.
- Author
-
Steven W Henning, Dinesh Jaishankar, Levi W Barse, Emilia R Dellacecca, Nicola Lancki, Kirsten Webb, Linda Janusek, Herbert L Mathews, Ronald N Price, and I Caroline Le Poole
- Subjects
Medicine ,Science - Abstract
Vitiligo is a T-cell mediated skin disorder characterized by progressive loss of skin color. In individuals genetically predisposed to the disease, various triggers contribute to the initiation of vitiligo. Precipitating factors can stress the skin, leading to T-cell activation and recruitment. Though hereditary factors are implicated in the pathogenesis of vitiligo, it is unknown whether precipitating, stressful events play a role in vitiligo. To understand this, we utilized a validated perceived stress scale (PSS) to measure this parameter in vitiligo patients compared to persons without vitiligo. Additionally, we probed a clinical database, using a knowledge linking software called ROCKET, to gauge stress-related conditions in the vitiligo patient population. From a pool of patients in an existing database, a hundred individuals with vitiligo and twenty-five age- and sex-matched comparison group of individuals without vitiligo completed an online survey to quantify their levels of perceived stress. In parallel, patients described specifics of their disease condition, including the affected body sites, the extent, duration and activity of their vitiligo. Perceived stress was significantly higher among vitiligo individuals compared to those without vitiligo. ROCKET analyses suggested signs of metabolic-related disease (i.e., 'stress') preceding vitiligo development. No correlation was found between perceived stress and the stage or the extent of disease, suggesting that elevated stress may not be a consequence of pigment loss alone. The data provide further support for stress as a precipitating factor in vitiligo development.
- Published
- 2020
- Full Text
- View/download PDF
9. Targeting Herpes Simplex Virus-1 gD by a DNA Aptamer Can Be an Effective New Strategy to Curb Viral Infection
- Author
-
Tejabhiram Yadavalli, Alex Agelidis, Dinesh Jaishankar, Kyle Mangano, Neel Thakkar, Kumar Penmetcha, and Deepak Shukla
- Subjects
DNA aptamer ,topical treatment ,herpes simplex virus 1 ,prophylaxis ,therapy ,Therapeutics. Pharmacology ,RM1-950 - Abstract
Herpes simplex virus type 1 (HSV-1) is an important factor for vision loss in developed countries. A challenging aspect of the ocular infection by HSV-1 is that common treatments, such as acyclovir, fail to provide effective topical remedies. Furthermore, it is not very clear whether the viral glycoproteins, required for HSV-1 entry into the host, can be targeted for an effective therapy against ocular herpes in vivo. Here, we demonstrate that HSV-1 envelope glycoprotein gD, which is essential for viral entry and spread, can be specifically targeted by topical applications of a small DNA aptamer to effectively control ocular infection by the virus. Our 45-nt-long DNA aptamer showed high affinity for HSV-1 gD (binding affinity constant [Kd] = 50 nM), which is strong enough to disrupt the binding of gD to its cognate host receptors. Our studies showed significant restriction of viral entry and replication in both in vitro and ex vivo studies. In vivo experiments in mice also resulted in loss of ocular infection under prophylactic treatment and statistically significant lower infection under therapeutic modality compared to random DNA controls. Thus, our studies validate the possibility that targeting HSV-1 entry glycoproteins, such as gD, can locally reduce the spread of infection and define a novel DNA aptamer-based approach to control HSV-1 infection of the eye.
- Published
- 2017
- Full Text
- View/download PDF
10. Viral Activation of Heparanase Drives Pathogenesis of Herpes Simplex Virus-1
- Author
-
Alex M. Agelidis, Satvik R. Hadigal, Dinesh Jaishankar, and Deepak Shukla
- Subjects
cornea ,cytokines ,heparan sulfate ,heparanase ,herpes simplex virus ,inflammation ,interferon ,ophthalmology ,transcription factors ,wound healing ,Biology (General) ,QH301-705.5 - Abstract
Herpes simplex virus-1 (HSV-1) causes lifelong recurrent pathologies without a cure. How infection by HSV-1 triggers disease processes, especially in the immune-privileged avascular human cornea, remains a major unresolved puzzle. It has been speculated that a cornea-resident molecule must tip the balance in favor of pro-inflammatory and pro-angiogenic conditions observed with herpetic, as well as non-herpetic, ailments of the cornea. Here, we demonstrate that heparanase (HPSE), a host enzyme, is the molecular trigger for multiple pathologies associated with HSV-1 infection. In human corneal epithelial cells, HSV-1 infection upregulates HPSE in a manner dependent on HSV-1 infected cell protein 34.5. HPSE then relocates to the nucleus to regulate cytokine production, inhibits wound closure, enhances viral spread, and thus generates a toxic local environment. Overall, our findings implicate activated HPSE as a driver of viral pathogenesis and call for further attention to this host protein in infection and other inflammatory disorders.
- Published
- 2017
- Full Text
- View/download PDF
11. Genital Herpes: Insights into Sexually Transmitted Infectious Disease
- Author
-
Dinesh Jaishankar and Deepak Shukla
- Subjects
herpes simplex virus ,virus entry ,viral glycoproteins ,viral latency ,antivirals ,Biology (General) ,QH301-705.5 - Abstract
Etiology, transmission and protection: Herpes simplex virus-2 (HSV-2) is a leading cause of sexually transmitted infections with recurring manifestations throughout the lifetime of infected hosts. Currently no effective vaccines or prophylactics exist that provide complete protection or immunity from the virus, which is endemic throughout the world. Pathology/Symptomatology: Primary and recurrent infections result in lesions and inflammation around the genital area and the latter accounts for majority of genital herpes instances. Immunocompromised patients including neonates are susceptible to additional systemic infections including debilitating consequences of nervous system inflammation. Epidemiology, incidence and prevalence: More than 500 million people are infected worldwide and most reported cases involve the age groups between 16-40 years, which coincides with an increase in sexual activity among this age group. While these numbers are an estimate, the actual numbers may be underestimated as many people are asymptomatic or do not report the symptoms. Treatment and curability: Currently prescribed medications, mostly nucleoside analogs, only reduce the symptoms caused by an active infection, but do not eliminate the virus or reduce latency. Therefore, no cure exists against genital herpes and infected patients suffer from periodic recurrences of disease symptoms for their entire lives. Molecular mechanisms of infection: The last few decades have generated many new advances in our understanding of the mechanisms that drive HSV infection. The viral entry receptors such as nectin-1 and HVEM have been identified, cytoskeletal signaling and membrane structures such as filopodia have been directly implicated in viral entry, host motor proteins and their viral ligands have been shown to facilitate capsid transport and many host and HSV proteins have been identified that help with viral replication and pathogenesis. New understanding has emerged on the role of autophagy and other innate immune mechanisms that are subverted to enhance HSV pathogenesis. This review summarizes our current understanding of HSV-2 and associated diseases and available or upcoming new treatments.
- Published
- 2016
- Full Text
- View/download PDF
12. Sulfotransferase and Heparanase: Remodeling Engines in Promoting Virus Infection and Disease Development
- Author
-
Dominik D. Kaltenbach, Dinesh Jaishankar, Meng Hao, Jacob C. Beer, Michael V. Volin, Umesh R. Desai, and Vaibhav Tiwari
- Subjects
heparan sulfate ,herpes simplex virus ,heparanse ,sulfotranferases ,heparan mimetic ,viral entry ,Therapeutics. Pharmacology ,RM1-950 - Abstract
An extraordinary binding site generated in heparan sulfate (HS) structures, during its biosynthesis, provides a unique opportunity to interact with multiple protein ligands including viral proteins, and therefore adds tremendous value to this master molecule. An example of such a moiety is the sulfation at the C3 position of glucosamine residues in HS chain via 3-O sulfotransferase (3-OST) enzymes, which generates a unique virus-cell fusion receptor during herpes simplex virus (HSV) entry and spread. Emerging evidence now suggests that the unique patterns in HS sulfation assist multiple viruses in invading host cells at various steps of their life cycles. In addition, sulfated-HS structures are known to assist in invading host defense mechanisms and initiating multiple inflammatory processes; a critical event in the disease development. All these processes are detrimental for the host and therefore raise the question of how HS-sulfation is regulated. Epigenetic modulations have been shown to be implicated in these reactions during HSV infection as well as in HS modifying enzyme sulfotransferases, and therefore pose a critical component in answering it. Interestingly, heparanase (HPSE) activity is shown to be upregulated during virus infection and multiple other diseases assisting in virus replication to promote cell and tissue damage. These phenomena suggest that sulfotransferases and HPSE serve as key players in extracellular matrix remodeling and possibly generating unique signatures in a given disease. Therefore, identifying the epigenetic regulation of OST genes, and HPSE resulting in altered yet specific sulfation patterns in HS chain during virus infection, will be a significant a step toward developing potential diagnostic markers and designing novel therapies.
- Published
- 2018
- Full Text
- View/download PDF
13. Emerging Roles of Heparanase in Viral Pathogenesis
- Author
-
Neel Thakkar, Tejabhiram Yadavalli, Dinesh Jaishankar, and Deepak Shukla
- Subjects
herpes simplex virus ,heparan sulfate ,heparanase ,Medicine - Abstract
Heparan sulfate (HS) is ubiquitously expressed on mammalian cells. It is a polysaccharide that binds growth factors, cytokines, and chemokines, and thereby controls several important physiological functions. Ironically, many human pathogens including viruses interact with it for adherence to host cells. HS functions can be regulated by selective modifications and/or selective cleavage of the sugar chains from the cell surface. In mammals, heparanase (HPSE) is the only known enzyme capable of regulating HS functions via a selective endoglycosidase activity that cleaves polymeric HS chains at internal sites. During homeostasis, HPSE expression and its endoglycosidase activity are tightly regulated; however, under stress conditions, including infection, its expression may be upregulated, which could contribute directly to the onset of several disease pathologies. Here we focus on viral infections exemplified by herpes simplex virus, dengue virus, human papillomavirus, respiratory syncytial virus, adenovirus, hepatitis C virus, and porcine respiratory and reproductive syncytial virus to summarize recent advances in understanding the highly significant, but emerging roles, of the enzyme HPSE in viral infection, spread and pathogenesis.
- Published
- 2017
- Full Text
- View/download PDF
14. Nanotherapeutics in transplantation: How do we get to clinical implementation?
- Author
-
Leah Plumblee, Carl Atkinson, Dinesh Jaishankar, Evan Scott, Gregory T. Tietjen, and Satish N. Nadig
- Subjects
Graft Rejection ,Immunosuppression Therapy ,Transplantation ,Immune Tolerance ,Humans ,Immunology and Allergy ,Pharmacology (medical) ,Organ Transplantation ,Immunosuppressive Agents - Abstract
Patients undergoing organ transplantation transition from one life-altering issue (organ dysfunction) to a lifelong commitment-immunosuppression. Regimens of immunosuppressive agents (ISAs) come with significant side effects and comorbidities. Recently, the use of nanoparticles (NPs) as a solution to the problems associated with the long-term and systemic use of ISAs in transplantation has emerged. This minireview describes the role of NPs in organ transplantation and discusses obstacles to clinical implementation and pathways to clinical translation.
- Published
- 2022
- Full Text
- View/download PDF
15. Mitochondrial responses to brain death in solid organ transplant
- Author
-
Meredith E. Taylor, Dinesh Jaishankar, Jessie W. Ho, Hasan B. Alam, Ankit Bharat, and Satish N. Nadig
- Abstract
Mitochondrial dynamics are central to the pathophysiology of cellular damage and inflammatory responses. In the context of solid organ transplantation, mitochondria are implicated in immune activation in donor organs that occurs after brain death, as they are critical to the regulation of cellular stress response, cell death, and display energetic adaptations through the adjustment of respiratory capacity depending on the cellular milieu. Mitochondrial damage activates mitochondrial systems of fission, fusion, biogenesis, and mitochondrial autophagy, or mitophagy. The mechanistic pathways as well as therapies targeting mitochondrial physiology have been studied as plausible ways to mitigate the negative effects of brain death on donor organs, though there is no summative evaluation of the multiple efforts across the field. This mini-review aims to discuss the interplay of donor brain death, mitochondrial dynamics, and impact on allograft function as it pertains to heart, lung, liver, and kidney transplants.
- Published
- 2023
- Full Text
- View/download PDF
16. Adoptive T-Cell Transfer to Treat Lymphangioleiomyomatosis
- Author
-
Fei Han, Christian M. Ankney, Daniel F. Dilling, Levi Barse, Alexander Yemelyanov, Steven W. Henning, I. Caroline Le Poole, Emilia R. Dellacecca, Dinesh Jaishankar, Vera P. Krymskaya, and Cormac Cosgrove
- Subjects
0301 basic medicine ,Pulmonary and Respiratory Medicine ,Adoptive cell transfer ,Lung Neoplasms ,T-Lymphocytes ,Transgene ,T cell ,Clinical Biochemistry ,Biology ,03 medical and health sciences ,0302 clinical medicine ,medicine ,Humans ,Lymphangioleiomyomatosis ,Cytotoxicity ,Molecular Biology ,Neuroendocrine cell ,Original Research ,T-cell receptor ,Cell Biology ,medicine.disease ,Adoptive Transfer ,030104 developmental biology ,medicine.anatomical_structure ,030228 respiratory system ,Cell culture ,Cancer research - Abstract
Patients with lymphangioleiomyomatosis (LAM) develop pulmonary cysts associated with neoplastic, smooth muscle–like cells that feature neuroendocrine cell markers. The disease preferentially affects premenopausal women. Existing therapeutics do not cure LAM. As gp100 is a diagnostic marker expressed by LAM lesions, we proposed to target this immunogenic glycoprotein using TCR transgenic T cells. To reproduce the genetic mutations underlying LAM, we cultured Tsc2(−/−) kidney tumor cells from aged Tsc2 heterozygous mice and generated a stable gp100-expressing cell line by lentiviral transduction. T cells were isolated from major histocompatibility complex–matched TCR transgenic pmel-1 mice to measure cytotoxicity in vitro, and 80% cytotoxicity was observed within 48 hours. Antigen-specific cytotoxicity was likewise observed using pmel-1 TCR-transduced mouse T cells, suggesting that transgenic T cells may likewise be useful to treat LAM in vivo. On intravenous injection, slow-growing gp100(+) LAM-like cells formed lung nodules that were readily detectable in severe combined immunodeficient/beige mice. Adoptive transfer of gp100-reactive but not wild-type T cells into mice significantly shrunk established lung tumors, even in the absence of anti–PD-1 therapy. These results demonstrate the treatment potential of adoptively transferred T cells to eliminate pulmonary lesions in LAM.
- Published
- 2020
- Full Text
- View/download PDF
17. Benign tumors in TSC are amenable to treatment by GD3 CAR T cells in mice
- Author
-
Emilia R. Dellacecca, Zhussipbek Mukhatayev, Suhail Akhtar, Levi Barse, Jesus Zamora-Pineda, Dinesh Jaishankar, Fei Han, Nikhilesh Gupta, Ancy Thomas, Thomas N. Darling, Daniel F. Dilling, Steven W. Henning, Saurav Sumughan, Agnes Lo, Cristina C. Vaca, Prathyaya Ramesh, I. Caroline Le Poole, Richard P. Junghans, Maria M. Picken, Joel Moss, Stephanie R. Zack, Denise M. Scholtens, Jasmine O. Zahid, Nicola Lancki, and Rohan Shivde
- Subjects
congenital, hereditary, and neonatal diseases and abnormalities ,medicine.medical_treatment ,Immunology ,T cells ,mTORC1 ,Therapeutics ,Immunotherapy, Adoptive ,Tuberous sclerosis ,Mice ,Immune system ,Antigen ,Tuberous Sclerosis ,medicine ,Cytotoxic T cell ,Animals ,Humans ,business.industry ,General Medicine ,Immunotherapy ,medicine.disease ,Chimeric antigen receptor ,nervous system diseases ,medicine.anatomical_structure ,Cancer research ,lipids (amino acids, peptides, and proteins) ,Female ,TSC1 ,business ,Research Article - Abstract
Mutations underlying disease in tuberous sclerosis complex (TSC) give rise to tumors with biallelic mutations in TSC1 or TSC2 and hyperactive mammalian target of rapamycin complex 1 (mTORC1). Benign tumors might exhibit de novo expression of immunogens, targetable by immunotherapy. As tumors may rely on ganglioside D3 (GD3) expression for mTORC1 activation and growth, we compared GD3 expression in tissues from patients with TSC and controls. GD3 was overexpressed in affected tissues from patients with TSC and also in aging Tsc2+/- mice. As GD3 overexpression was not accompanied by marked natural immune responses to the target molecule, we performed preclinical studies with GD3 chimeric antigen receptor (CAR) T cells. Polyfunctional CAR T cells were cytotoxic toward GD3-overexpressing targets. In mice challenged with Tsc2-/- tumor cells, CAR T cells substantially and durably reduced the tumor burden, correlating with increased T cell infiltration. We also treated aged Tsc2+/- heterozygous (>60 weeks) mice that carry spontaneous Tsc2-/- tumors with GD3 CAR or untransduced T cells and evaluated them at endpoint. Following CAR T cell treatment, the majority of mice were tumor free while all control animals carried tumors. The outcomes demonstrate a strong treatment effect and suggest that targeting GD3 can be successful in TSC.
- Published
- 2021
18. ERK1/2 phosphorylation predicts survival following anti-PD-1 immunotherapy in recurrent glioblastoma
- Author
-
Víctor A. Arrieta, Bin Zhang, Daniel J. Brat, Xiaoyang Ling, Daniel Zhang, Jinzhou Yuan, David Cieremans, Timothy F. Cloughesy, Lee Cooper, Peter Canoll, Robert M. Prins, Matthew McCord, Roger Stupp, Christina Amidei, Adam M. Sonabend, Gerson Rothschild, Catalina Lee-Chang, Li Chen, Cynthia Kassab, Pavan S. Upadhyayula, Jonathan T. Yamaguchi, Wenting Zhao, Dinesh Jaishankar, Kirsten Bell Burdett, Brice Laffleur, Amy B. Heimberger, Seong Jae Kang, Hui Zhang, Uttiya Basu, Andrew X. Chen, J. Robert Kane, Craig Horbinski, Peter A. Sims, Crismita Dmello, Jared K. Burks, Joseph Shilati, Jeffrey N. Bruce, Fabio M. Iwamoto, Raul Rabadan, Andrew Gould, Rimas V. Lukas, Junfei Zhao, Universidad Nacional Autónoma de México (UNAM), Columbia University [New York], Northwestern University Feinberg School of Medicine, The University of Texas M.D. Anderson Cancer Center [Houston], Microenvironment, Cell Differentiation, Immunology and Cancer (MICMAC), Université de Rennes 1 (UR1), Université de Rennes (UNIV-RENNES)-Université de Rennes (UNIV-RENNES)-Institut National de la Santé et de la Recherche Médicale (INSERM)-Structure Fédérative de Recherche en Biologie et Santé de Rennes ( Biosit : Biologie - Santé - Innovation Technologique ), David Geffen School of Medicine [Los Angeles], University of California [Los Angeles] (UCLA), University of California-University of California, NIHUnited States Department of Health and Human ServicesNational Institutes of Health (NIH) - USA [1R01NS110703-01A1, 5DP5OD02135605, R01-CA120813, 1R01-CA237418], SPORE for Translational Approaches to Brain Cancer [P50CA221747], Robert H. Lurie Cancer Center Support Grant [P30CA060553], Vagelos Precision Medicine Award [U54CA193313, U54CA209997, R35CA253126], Keep Punching, William Rhodes and Louise Tilzer-Rhodes Center for Glioblastoma at New York-Presbyterian Hospital, NIAIDUnited States Department of Health and Human ServicesNational Institutes of Health (NIH) - USANIH National Institute of Allergy and Infectious Diseases (NIAID) [1R01AI099195, R01AI134988], Mexican government through the Mexican National Council for Science and Technology, Plan of Combined Studies in Medicine of the National Autonomous University of Mexico, Medical Scientist Training ProgramUnited States Department of Health and Human ServicesNational Institutes of Health (NIH) - USA [T32GM007367], Cancer Center Support Grant [NCI CA060553], NIH through MD Andersons Cancer Center Support GrantUnited States Department of Health and Human ServicesNational Institutes of Health (NIH) - USA [CA016672], NCIUnited States Department of Health and Human ServicesNational Institutes of Health (NIH) - USANIH National Cancer Institute (NCI) [R50 CA23707], Structure Fédérative de Recherche en Biologie et Santé de Rennes ( Biosit : Biologie - Santé - Innovation Technologique )-Institut National de la Santé et de la Recherche Médicale (INSERM)-Université de Rennes 1 (UR1), Université de Rennes (UNIV-RENNES)-Université de Rennes (UNIV-RENNES), Universidad Nacional Autónoma de México = National Autonomous University of Mexico (UNAM), Université de Rennes (UR)-Institut National de la Santé et de la Recherche Médicale (INSERM)-Structure Fédérative de Recherche en Biologie et Santé de Rennes ( Biosit : Biologie - Santé - Innovation Technologique ), and University of California (UC)-University of California (UC)
- Subjects
MAPK/ERK pathway ,Cancer Research ,Myeloid ,MAP Kinase Signaling System ,medicine.medical_treatment ,[SDV]Life Sciences [q-bio] ,Article ,03 medical and health sciences ,0302 clinical medicine ,Medicine ,Humans ,Phosphorylation ,030304 developmental biology ,0303 health sciences ,Microglia ,business.industry ,Immunotherapy ,3. Good health ,Blockade ,medicine.anatomical_structure ,Oncology ,030220 oncology & carcinogenesis ,Cancer research ,Immunohistochemistry ,Biomarker (medicine) ,Neoplasm Recurrence, Local ,business ,Glioblastoma - Abstract
International audience; In two cohorts of patients with glioblastoma who received anti-PD-1, Sonabend and colleagues show that ERK1/2 phosphorylation, detected by immunohistochemistry, provides a biomarker for MAPK/ERK pathway activity and better survival on this therapy. Only a subset of recurrent glioblastoma (rGBM) responds to anti-PD-1 immunotherapy. Previously, we reported enrichment of BRAF/PTPN11 mutations in 30% of rGBM that responded to PD-1 blockade. Given that BRAF and PTPN11 promote MAPK/ERK signaling, we investigated whether activation of this pathway is associated with response to PD-1 inhibitors in rGBM, including patients that do not harbor BRAF/PTPN11 mutations. Here we show that immunohistochemistry for ERK1/2 phosphorylation (p-ERK), a marker of MAPK/ERK pathway activation, is predictive of overall survival following adjuvant PD-1 blockade in two independent rGBM patient cohorts. Single-cell RNA-sequencing and multiplex immunofluorescence analyses revealed that p-ERK was mainly localized in tumor cells and that high-p-ERK GBMs contained tumor-infiltrating myeloid cells and microglia with elevated expression of MHC class II and associated genes. These findings indicate that ERK1/2 activation in rGBM is predictive of response to PD-1 blockade and is associated with a distinct myeloid cell phenotype.
- Published
- 2021
- Full Text
- View/download PDF
19. Disruption of innate defense responses by endoglycosidase HPSE promotes cell survival
- Author
-
Benjamin A. Turturice, Satvik Hadigal, David Gonzalez, David L. Perkins, Anaamika Campeau, Alex Agelidis, Joshua Ames, Israel Vlodavsky, Deepak Shukla, Tejabhiram Yadavalli, Dinesh Jaishankar, Jacob M. Wozniak, Jin-Ping Li, James Hopkins, Lulia Koujah, Patricia W. Finn, Evan J. Kyzar, Chandrashekhar D. Patil, and Rahul K. Suryawanshi
- Subjects
Male ,0301 basic medicine ,Cell biology ,Programmed cell death ,Cell Survival ,Cell- och molekylärbiologi ,Defence mechanisms ,Herpesvirus 1, Human ,Biology ,Microbiology ,Extracellular matrix ,03 medical and health sciences ,0302 clinical medicine ,Transcription (biology) ,Transcriptional regulation ,Animals ,Heparanase ,Glucuronidase ,Inflammation ,Mice, Knockout ,Innate immunity ,Innate immune system ,Herpes Simplex ,Cellular immune response ,General Medicine ,Immunity, Innate ,Mice, Inbred C57BL ,030104 developmental biology ,030220 oncology & carcinogenesis ,Host-Pathogen Interactions ,Interferon Type I ,Necroptosis ,Medicine ,Female ,Transcription ,Reprogramming ,Cell and Molecular Biology ,Transcription Factors ,Research Article - Abstract
The drive to withstand environmental stresses and defend against invasion is a universal trait extant in all forms of life. While numerous canonical signaling cascades have been characterized in detail, it remains unclear how these pathways interface to generate coordinated responses to diverse stimuli. To dissect these connections, we followed heparanase (HPSE), a protein best known for its endoglycosidic activity at the extracellular matrix but recently recognized to drive various forms of late-stage disease through unknown mechanisms. Using herpes simplex virus-1 (HSV-1) infection as a model cellular perturbation, we demonstrate that HPSE acts beyond its established enzymatic role to restrict multiple forms of cell-intrinsic defense and facilitate host cell reprogramming by the invading pathogen. We reveal that cells devoid of HPSE are innately resistant to infection and counteract viral takeover through multiple amplified defense mechanisms. With a unique grasp of the fundamental processes of transcriptional regulation and cell death, HPSE represents a potent cellular intersection with broad therapeutic potential.
- Published
- 2021
- Full Text
- View/download PDF
20. A Palette of Cytokines to Measure Anti-Tumor Efficacy of T Cell-Based Therapeutics
- Author
-
I. Caroline Le Poole, Rohan Shivde, Dinesh Jaishankar, Prathyaya Ramesh, and Diana Saleiro
- Subjects
0301 basic medicine ,Cancer Research ,tumor ,immune monitoring ,medicine.medical_treatment ,T cell ,Review ,lcsh:RC254-282 ,03 medical and health sciences ,0302 clinical medicine ,Immune system ,Immunity ,medicine ,Tumor microenvironment ,business.industry ,Immunotherapy ,lcsh:Neoplasms. Tumors. Oncology. Including cancer and carcinogens ,cytokines ,Crosstalk (biology) ,030104 developmental biology ,Cytokine ,medicine.anatomical_structure ,Oncology ,polyfunctionality ,030220 oncology & carcinogenesis ,Cancer research ,immunotherapy ,business ,CD8 ,effector function - Abstract
Simple Summary Cytokines are molecules that help cells communicate at short range. Often, a single cytokine is measured in patient tissues to understand whether immune cells are actively clearing a tumor. We can learn whether a therapy works, even before changes in tumor burden are found. However, measuring multiple different cytokines better reflects ongoing anti-tumor activity. That is especially true for T cell-based therapies, as these cells secrete different cytokines when they eliminate tumors or not. We also need to consider that a single cytokine can perform activities that can either suppress, or support tumor growth. Taken together, we can best describe the function of T cells in a (pre) clinical setting by a palette of cytokines working together to paint a picture of ongoing immune responses to tumor cells. Abstract Cytokines are key molecules within the tumor microenvironment (TME) that can be used as biomarkers to predict the magnitude of anti-tumor immune responses. During immune monitoring, it has been customary to predict outcomes based on the abundance of a single cytokine, in particular IFN-γ or TGF-β, as a readout of ongoing anti-cancer immunity. However, individual cytokines within the TME can exhibit dual opposing roles. For example, both IFN-γ and TGF-β have been associated with pro- and anti-tumor functions. Moreover, cytokines originating from different cellular sources influence the crosstalk between CD4+ and CD8+ T cells, while the array of cytokines expressed by T cells is also instrumental in defining the mechanisms of action and efficacy of treatments. Thus, it becomes increasingly clear that a reliable readout of ongoing immunity within the TME will have to include more than the measurement of a single cytokine. This review focuses on defining a panel of cytokines that could help to reliably predict and analyze the outcomes of T cell-based anti-tumor therapies.
- Published
- 2021
21. 701 Nanoparticulate monobenzone (MBEH) as a potential drug candidate for melanoma
- Author
-
Steven W. Henning, Dinesh Jaishankar, Anqi Zhang, Basar Bilgicer, SonBinh Nhuyen, and I. Caroline Le Poole
- Subjects
Liposome ,medicine.diagnostic_test ,Tumor-infiltrating lymphocytes ,Chemistry ,Melanoma ,Melanocyte ,medicine.disease ,Flow cytometry ,Monobenzone ,medicine.anatomical_structure ,Depigmentation ,In vivo ,medicine ,Cancer research ,medicine.symptom ,medicine.drug - Abstract
Background Monobenzone (MBEH) is a skin depigmenting agent FDA-approved for topical applications.1 It specifically interacts with tyrosinase,2 a key enzyme in melanogenesis, to form reactive quinones that are toxic to pigmented cells, including melanoma cells. As melanoma cells express abundant tyrosinase activity,3 repurposing MBEH to target melanoma cells might serve as a treatment strategy. Furthermore, quinones can haptenize tyrosinase,4 supporting neo-antigen formation.5 Modified tumor antigens then initiate an immune cascade, engaging T cells to target tumor cells. This biphasic effect of MBEH makes it a suitable candidate to target melanoma. Although topical treatment of MBEH can suppress subcutaneous melanoma growth in vivo,6 systemic administration of the drug was toxic,7 limiting the application of MBEH for metastatic disease. To overcome this limitation, we encapsulated MBEH and its derivatives into nanoscale liposomes (~100 nm) and evaluated its anti-tumor efficacy. Methods Liposomes were prepared8 and MBEH was loaded into the liposomes. Loading was evaluated using mass spectroscopy and nuclear magnetic resonance (NMR) spectroscopy. In vitro cytotoxicity of liposomal MBEH to mouse and human melanoma cell lines was evaluated by MTT assays. Meanwhile, in vivo trafficking of fluorescent liposomes to B16-F10 tumors and vital organs was evaluated in tissue homogenates by flow cytometry. The anti-tumor effects of liposomal MBEH towards subcutaneously injected B16-F10 melanoma cells were evaluated in C57BL/6 mice (n=4 per group) over time. Results Mass spectroscopy and NMR data revealed that MBEH was encapsulated into the liposomes at 2.3%mol MBEH per liposome. Liposomal MBEH was toxic to both mouse and human melanoma cells with lower half maximal inhibitory concentration (IC50) values in B16-F10 and A375 and higher IC50 values in 888-A2 and 624.38 cells. In vivo trafficking in mice revealed ~ 55% uptake of liposomal fluorescence by the tumor. In contrast to empty liposomes (mean tumor volume on day 21: 467.5 mm3), MBEH loaded liposomes significantly (P Conclusions We provide a proof-of-concept to use nanoparticulate MBEH to target the melanogenic pathway in melanoma. A detailed study of MBEH loading, nanoparticle stability, and tumor infiltrating lymphocyte can further establish nanoparticulate MBEH as a potential drug candidate for melanoma. Acknowledgements This work was supported by a grant from the Sherman Fairchild Foundation. References Bolognia JL, Lapia K, and Somma S, Depigmentation therapy. Dermatologic Therapy 2001. 14(1): p. 29–34. McGuire J and Hendee J, Biochemical Basis for Depigmentation of Skin by Phenolic Germicides. Journal of Investigative Dermatology 1971. 57(4): p. 256–261. Michaeli Y, et al., Melanoma cells present high levels of HLA-A2-tyrosinase in association with instability and aberrant intracellular processing of tyrosinase. European Journal of Immunology, 2012. 42(4): p. 842–850. Manini P, et al., A reactive ortho-quinone generated by tyrosinase-catalyzed oxidation of the skin depigmenting agent monobenzone: self-coupling and thiol-conjugation reactions and possible implications for melanocyte toxicity. Chem Res Toxicol, 2009. 22(8): p. 1398–405. 5. Westerhof W, et al., The haptenation theory of vitiligo and melanoma rejection: a close-up. Experimental Dermatology 2011. 20(2): p. 92–96. Hariharan V, et al., Topical application of bleaching phenols; in-vivo studies and mechanism of action relevant to melanoma treatment. Melanoma Res, 2011. 21(2): p. 115–26. Kelly KH, Bierman HR, and Shimkin MB, Negative effects of oral monobenzyl ether of hydroquinone in malignant melanoma in man. proceedings of the society for experimental Biology and Medicine 1952. 79(4): p. 589–590. Stefanick JF, et al., A systematic analysis of peptide linker length and liposomal polyethylene glycol coating on cellular uptake of peptide-targeted liposomes. ACS Nano 2013. 7(4): p. 2935–2947.
- Published
- 2020
- Full Text
- View/download PDF
22. HSP70i
- Author
-
Dinesh, Jaishankar, Cormac, Cosgrove, Prathyaya, Ramesh, James, Mahon, Rohan, Shivde, Emilia R, Dellacecca, Shiayin F, Yang, Jeffrey, Mosenson, José A, Guevara-Patiño, and I Caroline, Le Poole
- Subjects
Male ,Original Paper ,Skin Neoplasms ,Autoimmunity ,DNA, Neoplasm ,Models, Biological ,Antibodies ,Cell Degranulation ,Killer Cells, Natural ,Mice, Inbred C57BL ,Cell Line, Tumor ,Mutation ,Animals ,Humans ,HSP70 Heat-Shock Proteins ,Melanoma - Abstract
Developing immunosuppressive therapies for autoimmune diseases comes with a caveat that immunosuppression may promote the risk of developing other conditions or diseases. We have previously shown that biolistic delivery of an expression construct encoding inducible HSP70 (HSP70i) with one amino acid modification in the dendritic cell (DC) activating moiety 435–445 (HSP70i(Q435A)) to mouse skin resulted in significant immunosuppressive activity of autoimmune vitiligo, associated with fewer tissue infiltrating T cells. To prepare HSP70i(Q435A) as a potential therapeutic for autoimmune vitiligo, in this study we evaluated whether and how biolistic delivery of HSP70i(Q435A) in mice affects anti-tumor responses. We found that HSP70i(Q435A) in fact supports anti-tumor responses in melanoma-challenged C57BL/6 mice. Biolistic delivery of the HSP70i(Q435A)-encoding construct to mice elicited significant anti-HSP70 titers, and anti-HSP70 IgG and IgM antibodies recognize surface-expressed and cytoplasmic HSP70i in human and mouse melanoma cells. A peptide scan revealed that the anti-HSP70 antibodies recognize a specific C-terminal motif within the HSP70i protein. The antibodies elicited surface CD107A expression among mouse NK cells, representative of antibody-mediated cellular cytotoxicity (ADCC), supporting the concept, that HSP70i(Q435A)-encoding DNA elicits a humoral response to the stress protein expressed selectively on the surface of melanoma cells. Thus, besides limiting autoimmunity and inflammation, HSP70i(Q435A) elicits humoral responses that limit tumor growth and may be used in conjunction with immune checkpoint inhibitors to not only control tumor but to also limit adverse events following tumor immunotherapy. SUPPLEMENTARY INFORMATION: The online version contains supplementary material available at 10.1007/s12192-021-01229-x.
- Published
- 2020
23. Disruption of innate defense responses by endoglycosidase HPSE promotes cell survival
- Author
-
David L. Perkins, James Hopkins, Tejabhiram Yadavalli, Jin-Ping Li, Benjamin A. Turturice, Dinesh Jaishankar, Anaamika Campeau, Lulia Koujah, Evan J. Kyzar, Alex Agelidis, Chandrashekhar D. Patil, Israel Vlodavsky, Rahul K. Suryawanshi, Joshua Ames, Jacob M. Wozniak, Deepak Shukla, Patricia W. Finn, David Gonzalez, and Satvik Hadigal
- Subjects
Extracellular matrix ,Programmed cell death ,biology ,Transcriptional regulation ,biology.protein ,Defence mechanisms ,Heparanase ,Reprogramming ,Cell survival ,Endoglycosidase ,Cell biology - Abstract
The drive to withstand environmental stresses and defend against invasion is a universal trait extant in all forms of life. While numerous canonical signaling cascades have been characterized in detail, it remains unclear how these pathways interface to generate coordinated responses to diverse stimuli. To dissect these connections, we follow heparanase (HPSE), a protein best known for its endoglycosidic activity at the extracellular matrix but recently recognized to drive various forms of late stage disease through unknown mechanisms. Using herpes simplex virus-1 (HSV-1) infection as a model cellular perturbation, we demonstrate that HPSE acts beyond its established enzymatic role to restrict multiple forms of cell-intrinsic defense and facilitate host cell reprogramming by the invading pathogen. We reveal that cells devoid of HPSE are innately resistant to infection and counteract viral takeover through multiple amplified defense mechanisms. With a unique grasp of the fundamental processes of transcriptional regulation and cell death, HPSE represents a potent cellular intersection with broad therapeutic potential.
- Published
- 2020
- Full Text
- View/download PDF
24. A Rapid Method for Multispectral Fluorescence Imaging of Frozen Tissue Sections
- Author
-
Dinesh Jaishankar, I. Caroline Le Poole, Ryan Deaton, and Cormac Cosgrove
- Subjects
Fluorescence-lifetime imaging microscopy ,Tissue Fixation ,Fluorophore ,General Immunology and Microbiology ,Chemistry ,General Chemical Engineering ,General Neuroscience ,Multispectral image ,Tissue sample ,Fluorescence ,Article ,General Biochemistry, Genetics and Molecular Biology ,Staining ,Mice ,chemistry.chemical_compound ,Multiple markers ,Animals ,Frozen Sections ,Humans ,Analysis software ,Frozen tissue ,Biomedical engineering - Abstract
Multispectral fluorescence imaging on formalin-fixed paraffin-embedded (FFPE) tissues enables the detection of multiple markers in a single tissue sample that can provide information about antigen coexpression and spatial distribution of the markers. However, a lack of suitable antibodies for formalin-fixed tissues may restrict the nature of markers that can be detected. In addition, the staining method is time-consuming. Here we describe a rapid method to perform multispectral fluorescence imaging on frozen tissues. The method includes the fluorophore combinations used, detailed steps for the staining of mouse and human frozen tissues, and the scanning, acquisition, and analysis procedures. For staining analysis, a commercially available semiautomated multispectral fluorescence imaging system is used. Through this method, up to six different markers were stained and detected in a single frozen tissue section. The machine learning analysis software can phenotype cells that can be used for quantitative analysis. The method described here for frozen tissues is useful for the detection of markers that cannot be detected in FFPE tissues or for which antibodies are not available for FFPE tissues.
- Published
- 2020
- Full Text
- View/download PDF
25. The relationship between stress and vitiligo: Evaluating perceived stress and electronic medical record data
- Author
-
Levi Barse, Nicola Lancki, Steven W. Henning, Dinesh Jaishankar, Herbert L. Mathews, I. Caroline Le Poole, Ron Price, Kirsten Webb, Emilia R. Dellacecca, and Linda Witek Janusek
- Subjects
0301 basic medicine ,Male ,Questionnaires ,T-Lymphocytes ,Perceived Stress Scale ,Social Sciences ,Vitiligo ,Disease ,Epithelium ,Heat Shock Response ,030207 dermatology & venereal diseases ,0302 clinical medicine ,Drug Metabolism ,Animal Cells ,Surveys and Questionnaires ,Stress (linguistics) ,Medicine and Health Sciences ,Psychology ,Young adult ,Child ,skin and connective tissue diseases ,Cellular Stress Responses ,Metabolic Syndrome ,Multidisciplinary ,integumentary system ,Electronic medical record ,Middle Aged ,Research Design ,Cell Processes ,Child, Preschool ,Melanocytes ,Medicine ,Female ,Cellular Types ,Anatomy ,Research Article ,Adult ,medicine.medical_specialty ,Adolescent ,Patients ,Science ,Immunology ,Psychological Stress ,Research and Analysis Methods ,Autoimmune Diseases ,03 medical and health sciences ,Young Adult ,Stress, Physiological ,Mental Health and Psychiatry ,medicine ,Humans ,Pharmacokinetics ,Chromatophores ,Aged ,Pharmacology ,Survey Research ,business.industry ,Infant, Newborn ,Infant ,Biology and Life Sciences ,Epithelial Cells ,Cell Biology ,medicine.disease ,Dermatology ,Health Care ,030104 developmental biology ,Biological Tissue ,Skin color ,Metabolic Disorders ,Clinical Immunology ,Metabolic syndrome ,Clinical Medicine ,business ,Heat-Shock Response ,Stress, Psychological - Abstract
Vitiligo is a T-cell mediated skin disorder characterized by progressive loss of skin color. In individuals genetically predisposed to the disease, various triggers contribute to the initiation of vitiligo. Precipitating factors can stress the skin, leading to T-cell activation and recruitment. Though hereditary factors are implicated in the pathogenesis of vitiligo, it is unknown whether precipitating, stressful events play a role in vitiligo. To understand this, we utilized a validated perceived stress scale (PSS) to measure this parameter in vitiligo patients compared to persons without vitiligo. Additionally, we probed a clinical database, using a knowledge linking software called ROCKET, to gauge stress-related conditions in the vitiligo patient population. From a pool of patients in an existing database, a hundred individuals with vitiligo and twenty-five age- and sex-matched comparison group of individuals without vitiligo completed an online survey to quantify their levels of perceived stress. In parallel, patients described specifics of their disease condition, including the affected body sites, the extent, duration and activity of their vitiligo. Perceived stress was significantly higher among vitiligo individuals compared to those without vitiligo. ROCKET analyses suggested signs of metabolic-related disease (i.e., 'stress') preceding vitiligo development. No correlation was found between perceived stress and the stage or the extent of disease, suggesting that elevated stress may not be a consequence of pigment loss alone. The data provide further support for stress as a precipitating factor in vitiligo development.
- Published
- 2020
26. Publisher Correction: ERK1/2 phosphorylation predicts survival following anti-PD-1 immunotherapy in recurrent glioblastoma
- Author
-
Víctor A. Arrieta, Andrew X. Chen, J. Robert Kane, Seong Jae Kang, Cynthia Kassab, Crismita Dmello, Junfei Zhao, Kirsten B. Burdett, Pavan S. Upadhyayula, Catalina Lee-Chang, Joseph Shilati, Dinesh Jaishankar, Li Chen, Andrew Gould, Daniel Zhang, Jinzhou Yuan, Wenting Zhao, Xiaoyang Ling, Jared K. Burks, Brice Laffleur, Christina Amidei, Jeffrey N. Bruce, Rimas V. Lukas, Jonathan T. Yamaguchi, David Cieremans, Gerson Rothschild, Uttiya Basu, Matthew McCord, Daniel J. Brat, Hui Zhang, Lee A. D. Cooper, Bin Zhang, Peter Sims, Tim F. Cloughesy, Robert Prins, Peter Canoll, Roger Stupp, Amy B. Heimberger, Craig Horbinski, Fabio M. Iwamoto, Raul Rabadan, and Adam M. Sonabend
- Subjects
Cancer Research ,Oncology - Published
- 2022
- Full Text
- View/download PDF
27. BIOM-31. ERK1/2 PHOSPHORYLATION PREDICTS SURVIVAL FOLLOWING ANTI-PD-1 IMMUNOTHERAPY IN RECURRENT GLIOBLASTOMA
- Author
-
Raul Rabadan, Jeffrey N. Bruce, Peter A. Sims, Daniel J. Brat, Matthew R McCord, Pavan S. Upadhyayula, Andrew Gould, Hui Zhang, Gerson Rothschild, Xiaoyang Ling, Kirsten Bell Burdett, Fabio M. Iwamoto, Lee Cooper, Joseph Shilati, Rimas V. Lukas, Junfei Zhao, Cynthia Kassab, Uttiya Basu, Brice Laffleur, Wenting Zhao, Dinesh Jaishankar, Li Chen, Daniel Zhang, Peter Canoll, Robert M. Prins, Amy B. Heimberger, J. Robert Kane, Timothy F. Cloughesy, Catalina Chang, Crismita Dmello, Jared K. Burks, Craig Horbinski, Jinzhou Yuan, David Cieremans, Jonathan T. Yamaguchi, Andrew X. Chen, Roger Stupp, Christina Amidei, Víctor A. Arrieta, Bin Zhang, Adam M. Sonabend, and Seong Jae Kang
- Subjects
Cancer Research ,Cell cycle checkpoint ,Extracellular matrix-cell signaling ,Microglia ,business.industry ,medicine.medical_treatment ,Cancer ,Immunotherapy ,medicine.disease ,medicine.anatomical_structure ,Oncology ,medicine ,Cancer research ,Phosphorylation ,Immunohistochemistry ,Neurology (clinical) ,Signal transduction ,business - Abstract
PD-1 checkpoint inhibition has led to remarkable clinical responses in several cancer types. Whereas PD-1 blockade has not shown an overall survival (OS) benefit for glioblastoma (GBM) patients, a subset of them exhibit long-term responses to this immunotherapy. Previously, we reported an enrichment of BRAF/PTPN11 activating mutations in 30% of recurrent GBMs that responded to PD-1 blockade, but the molecular profile of the majority of responders remained elusive. Given that BRAF and PTPN11 promote MAPK/ERK signaling, we investigated whether activation of this pathway is associated with response to PD-1 inhibitors in recurrent GBM, including patients that do not harbor BRAF/PTPN11 mutations. Immunohistochemistry for ERK1/2 phosphorylation (p-ERK), a marker of MAPK/ERK pathway activation, was performed in a discovery cohort including pre-treatment specimens of 29 recurrent GBM patients treated with adjuvant PD-1 blockade, and 33 patients who did not undergo immunotherapy. p-ERK was predictive of response and OS following PD-1 blockade. Yet p-ERK was not associated with OS in patients not treated with immunotherapy. p-ERK was also associated with OS in a validation GBM cohort treated with adjuvant anti-PD-1 therapy. Single-cell RNA-seq and multiplex-immunofluorescence analyses revealed that p-ERK was mainly localized in tumor cells and high p-ERK GBMs contained tumor-infiltrating myeloid cells and microglia with elevated expression of MHC class II and associated genes. Thus, our findings indicate that ERK1/2 activation in recurrent GBM is predictive of response to PD-1 blockade and is associated with a distinct myeloid cell phenotype.
- Published
- 2021
- Full Text
- View/download PDF
28. Targeting Herpes Simplex Virus-1 gD by a DNA Aptamer Can Be an Effective New Strategy to Curb Viral Infection
- Author
-
Deepak Shukla, Kumar K.R. Penmetcha, Neel Thakkar, Dinesh Jaishankar, Tejabhiram Yadavalli, Kyle Mangano, and Alex Agelidis
- Subjects
0301 basic medicine ,viruses ,Aptamer ,030106 microbiology ,Biology ,medicine.disease_cause ,Article ,Virus ,Ocular herpes ,03 medical and health sciences ,chemistry.chemical_compound ,Viral entry ,Drug Discovery ,medicine ,Receptor ,chemistry.chemical_classification ,therapy ,lcsh:RM1-950 ,DNA aptamer ,medicine.disease ,Virology ,3. Good health ,topical treatment ,lcsh:Therapeutics. Pharmacology ,030104 developmental biology ,Herpes simplex virus ,chemistry ,Molecular Medicine ,prophylaxis ,herpes simplex virus 1 ,Glycoprotein ,DNA - Abstract
Herpes simplex virus type 1 (HSV-1) is an important factor for vision loss in developed countries. A challenging aspect of the ocular infection by HSV-1 is that common treatments, such as acyclovir, fail to provide effective topical remedies. Furthermore, it is not very clear whether the viral glycoproteins, required for HSV-1 entry into the host, can be targeted for an effective therapy against ocular herpes in vivo. Here, we demonstrate that HSV-1 envelope glycoprotein gD, which is essential for viral entry and spread, can be specifically targeted by topical applications of a small DNA aptamer to effectively control ocular infection by the virus. Our 45-nt-long DNA aptamer showed high affinity for HSV-1 gD (binding affinity constant [Kd] = 50 nM), which is strong enough to disrupt the binding of gD to its cognate host receptors. Our studies showed significant restriction of viral entry and replication in both in vitro and ex vivo studies. In vivo experiments in mice also resulted in loss of ocular infection under prophylactic treatment and statistically significant lower infection under therapeutic modality compared to random DNA controls. Thus, our studies validate the possibility that targeting HSV-1 entry glycoproteins, such as gD, can locally reduce the spread of infection and define a novel DNA aptamer-based approach to control HSV-1 infection of the eye., Graphical Abstract
- Published
- 2017
- Full Text
- View/download PDF
29. Drug-encapsulated carbon (DECON): A novel platform for enhanced drug delivery
- Author
-
Deepak Shukla, Rahul K. Suryawanshi, Dinesh Jaishankar, Tejabhiram Yadavalli, Lulia Koujah, Neel Thakkar, James Hopkins, Alex Agelidis, and Joshua Ames
- Subjects
Drug ,medicine.drug_class ,media_common.quotation_subject ,Acyclovir ,02 engineering and technology ,CHO Cells ,Herpesvirus 1, Human ,Pharmacology ,Antiviral Agents ,Virus ,Corneal Diseases ,03 medical and health sciences ,Mice ,Cricetulus ,Cell Line, Tumor ,Highly porous ,Chlorocebus aethiops ,medicine ,Animals ,Humans ,Health and Medicine ,Vero Cells ,Research Articles ,030304 developmental biology ,media_common ,0303 health sciences ,Polymer-drug conjugates ,Drug Carriers ,Mice, Inbred BALB C ,Multidisciplinary ,Herpes Genitalis ,Chemistry ,SciAdv r-articles ,021001 nanoscience & nanotechnology ,In vitro ,Carbon ,3. Good health ,Mice, Inbred C57BL ,Disease Models, Animal ,Charcoal ,Drug delivery ,Female ,Antiviral drug ,0210 nano-technology ,Dosing Frequency ,HeLa Cells ,Research Article - Abstract
We describe a safe and natural carbon-based system to actively deliver drugs to mucosal surfaces, Current drug-delivery systems are designed primarily for parenteral applications and are either lipid or polymer drug conjugates. In our quest to inhibit herpes simplex virus infection via the compounds found in commonly used cosmetic products, we found that activated carbon particles inhibit infection and, in addition, substantially improve topical delivery and, hence, the efficacy of a common antiviral drug, acyclovir (ACV). Our in vitro studies demonstrate that highly porous carbon structures trapped virions, blocked infection and substantially improved efficacy when ACV was loaded onto them. Also, using murine models of corneal and genital herpes infections, we show that the topical use of drug-encapsulated carbon (DECON) reduced dosing frequency, shortened treatment duration, and exhibited higher therapeutic efficacy than currently approved topical or systemic antivirals alone. DECON is a nontoxic, cost-effective and nonimmunogenic alternative to current topical drug-delivery systems that is uniquely triggered for drug release by virus trapping.
- Published
- 2019
30. 549 Design and in vitro efficacy of TRP-1 CAR T cells to target melanoma
- Author
-
I. C. Le Poole, Rohan Shivde, Ancy Thomas, and Dinesh Jaishankar
- Subjects
Chemistry ,Melanoma ,Cancer research ,medicine ,Cell Biology ,Dermatology ,Car t cells ,medicine.disease ,Molecular Biology ,Biochemistry ,In vitro - Published
- 2021
- Full Text
- View/download PDF
31. Genital Herpes: Insights into Sexually Transmitted Infectious Disease
- Author
-
Deepak Shukla and Dinesh Jaishankar
- Subjects
0301 basic medicine ,Applied Microbiology ,viruses ,Disease ,virus entry ,medicine.disease_cause ,Biochemistry, Genetics and Molecular Biology (miscellaneous) ,Microbiology ,Applied Microbiology and Biotechnology ,Virus ,03 medical and health sciences ,antivirals ,Immunity ,Viral entry ,Virology ,viral latency ,Genetics ,Medicine ,lcsh:QH301-705.5 ,Molecular Biology ,Nucleoside analogue ,business.industry ,Cell Biology ,herpes simplex virus ,3. Good health ,030104 developmental biology ,Herpes simplex virus ,lcsh:Biology (General) ,Viral replication ,Infectious disease (medical specialty) ,Immunology ,Parasitology ,viral glycoproteins ,business ,medicine.drug - Abstract
Etiology, transmission and protection: Herpes simplex virus-2 (HSV-2) is a leading cause of sexually transmitted infections with recurring manifestations throughout the lifetime of infected hosts. Currently no effective vaccines or prophylactics exist that provide complete protection or immunity from the virus, which is endemic throughout the world. Pathology/Symptomatology: Primary and recurrent infections result in lesions and inflammation around the genital area and the latter accounts for majority of genital herpes instances. Immunocompromised patients including neonates are susceptible to additional systemic infections including debilitating consequences of nervous system inflammation. Epidemiology, incidence and prevalence: More than 500 million people are infected worldwide and most reported cases involve the age groups between 16-40 years, which coincides with an increase in sexual activity among this age group. While these numbers are an estimate, the actual numbers may be underestimated as many people are asymptomatic or do not report the symptoms. Treatment and curability: Currently prescribed medications, mostly nucleoside analogs, only reduce the symptoms caused by an active infection, but do not eliminate the virus or reduce latency. Therefore, no cure exists against genital herpes and infected patients suffer from periodic recurrences of disease symptoms for their entire lives. Molecular mechanisms of infection: The last few decades have generated many new advances in our understanding of the mechanisms that drive HSV infection. The viral entry receptors such as nectin-1 and HVEM have been identified, cytoskeletal signaling and membrane structures such as filopodia have been directly implicated in viral entry, host motor proteins and their viral ligands have been shown to facilitate capsid transport and many host and HSV proteins have been identified that help with viral replication and pathogenesis. New understanding has emerged on the role of autophagy and other innate immune mechanisms that are subverted to enhance HSV pathogenesis. This review summarizes our current understanding of HSV-2 and associated diseases and available or upcoming new treatments.
- Published
- 2016
- Full Text
- View/download PDF
32. Sulfotransferase and Heparanase: Remodeling Engines in Promoting Virus Infection and Disease Development
- Author
-
Jacob C. Beer, Dominik D. Kaltenbach, Dinesh Jaishankar, Vaibhav Tiwari, Meng Hao, Michael V. Volin, and Umesh R. Desai
- Subjects
0301 basic medicine ,sulfotranferases ,Sulfotransferase ,Review ,Biology ,Virus ,heparan mimetic ,03 medical and health sciences ,chemistry.chemical_compound ,heparanse ,Sulfation ,Viral entry ,Heparanase ,Pharmacology (medical) ,Epigenetics ,Pharmacology ,lcsh:RM1-950 ,Heparan sulfate ,herpes simplex virus ,Cell biology ,030104 developmental biology ,lcsh:Therapeutics. Pharmacology ,Viral replication ,chemistry ,viral entry ,heparan sulfate - Abstract
An extraordinary binding site generated in heparan sulfate (HS) structures, during its biosynthesis, provides a unique opportunity to interact with multiple protein ligands including viral proteins, and therefore adds tremendous value to this master molecule. An example of such a moiety is the sulfation at the C3 position of glucosamine residues in HS chain via 3-O sulfotransferase (3-OST) enzymes, which generates a unique virus-cell fusion receptor during herpes simplex virus (HSV) entry and spread. Emerging evidence now suggests that the unique patterns in HS sulfation assist multiple viruses in invading host cells at various steps of their life cycles. In addition, sulfated-HS structures are known to assist in invading host defense mechanisms and initiating multiple inflammatory processes; a critical event in the disease development. All these processes are detrimental for the host and therefore raise the question of how HS-sulfation is regulated. Epigenetic modulations have been shown to be implicated in these reactions during HSV infection as well as in HS modifying enzyme sulfotransferases, and therefore pose a critical component in answering it. Interestingly, heparanase (HPSE) activity is shown to be upregulated during virus infection and multiple other diseases assisting in virus replication to promote cell and tissue damage. These phenomena suggest that sulfotransferases and HPSE serve as key players in extracellular matrix remodeling and possibly generating unique signatures in a given disease. Therefore, identifying the epigenetic regulation of OST genes, and HPSE resulting in altered yet specific sulfation patterns in HS chain during virus infection, will be a significant a step toward developing potential diagnostic markers and designing novel therapies.
- Published
- 2018
- Full Text
- View/download PDF
33. An off-target effect of BX795 blocks herpes simplex virus type 1 infection of the eye
- Author
-
Neel Thakkar, Satvik Hadigal, Dinesh Jaishankar, Alex Agelidis, Joshua Ames, Abraam M. Yakoub, Tejabhiram Yadavalli, and Deepak Shukla
- Subjects
0301 basic medicine ,Swine ,viruses ,Mucocutaneous zone ,Herpesvirus 1, Human ,Thiophenes ,Eye ,medicine.disease_cause ,Antiviral Agents ,Article ,Ocular herpes ,Virus ,03 medical and health sciences ,TANK-binding kinase 1 ,medicine ,Animals ,Humans ,Mice, Inbred BALB C ,Nucleoside analogue ,business.industry ,Kinase ,Epithelium, Corneal ,Virion ,Herpes Simplex ,General Medicine ,medicine.disease ,Virology ,Epithelium ,Enzyme Activation ,Disease Models, Animal ,Pyrimidines ,030104 developmental biology ,medicine.anatomical_structure ,Herpes simplex virus ,business ,Proto-Oncogene Proteins c-akt ,medicine.drug - Abstract
Herpes simplex virus type-1 (HSV-1) causes recurrent mucocutaneous lesions in the eye that may advance to corneal blindness. Nucleoside analogs exemplified by acyclovir (ACV) form the primary class of anti-herpetic drugs but this class suffers limitations due to the emergence of viral resistance and other side effects. While studying the molecular basis of ocular HSV-1 infection, we observed that BX795, a commonly used inhibitor of TANK-binding kinase-1 (TBK1), strongly suppressed infection by multiple strains of HSV-1 in transformed and primary human cells, cultured human and animal corneas, and a murine model of ocular infection. Our investigations revealed that the antiviral activity of BX795 relies on targeting Akt phosphorylation in infected cells leading to the blockage of viral protein synthesis. This small molecule inhibitor, which could also be effective against ACV-resistant HSV-1 strains, shows promise as an alternative to existing drugs and as an effective topical therapy for ocular herpes infection. Collectively, our results obtained using multiple infection models and virus strains establish BX795 as a promising lead compound for broad-spectrum antiviral applications in humans.
- Published
- 2018
- Full Text
- View/download PDF
34. Herpesvirus-encoded microRNAs detected in human gingiva alter host cell transcriptome and regulate viral infection
- Author
-
Jennifer Shango, Dinesh Jaishankar, Afsar Raza Naqvi, Maria F. Brambila, Deepak Shukla, Gloria Martínez, Shirin Hasan, Alexandra Seal, Salvador Nares, Tejabhiram Yadavalli, and Gabriela Chapa
- Subjects
0301 basic medicine ,Gene Expression Regulation, Viral ,Keratinocytes ,Biophysics ,Gingiva ,Herpesvirus 1, Human ,Biology ,Biochemistry ,Article ,Transcriptome ,Pathogenesis ,03 medical and health sciences ,0302 clinical medicine ,Immune system ,Downregulation and upregulation ,Structural Biology ,Viral entry ,microRNA ,Genetics ,Humans ,Molecular Biology ,Gene ,Periodontal Diseases ,Innate immune system ,Binding Sites ,030206 dentistry ,Cell biology ,MicroRNAs ,030104 developmental biology ,Virus Diseases ,Herpesvirus 8, Human ,RNA, Viral - Abstract
MicroRNAs (miRNAs) are small, non-coding RNAs of ~18-25 nucleotides that have gained extensive attention as critical regulators in complex gene networks including immune cell lineage commitment, differentiation, maturation, and maintenance of immune homeostasis and function. Many viruses encode miRNAs that directly downregulate the expression of factors of the innate immune system, which includes proteins involved in promoting apoptosis and recruitment. In this study, we examined the expression profiles of three previously identified viral miRNAs (v-miRs) from the human herpesvirus (HHV) family, HSV-1 (miR-H1), KSHV (miR-K12-3-3p), and HCMV (miR-US4) in healthy and diseased periodontal tissues and observed increased levels of v-miRs in diseased tissues. To understand the significance of this increase, we overexpressed v-miRs in human oral keratinocytes (HOK), a common target for various HHV, and analyzed the impact of miR-H1 and miR-K12-3-3p on the host transcriptome. More than 1300 genes were altered in HOK overexpressing miR-H1 and miR-K12-3-3p. Global pathway analysis of deregulated genes identified several key cellular pathways that may favor viral persistence. Using bioinformatic analysis, we predicted hundreds of potential v-miR binding sites on genes downregulated by miR-H1 and miR-K12-3-3p and validated three novel target v-miR sites suggesting widespread direct and indirect modulation of numerous host genes/pathways by a single v-miR. Finally, in vitro HSV-1 infection assays showed that miR-H1 can regulate viral entry and infection in human oral keratinocytes (HOK). Overall, our results demonstrate clinical and functional relevance of pathogenic viral molecules viz., v-miRs that regulate both host and viral functions and may contribute to the pathogenesis of inflammatory oral diseases.
- Published
- 2017
35. Emerging Roles of Heparanase in Viral Pathogenesis
- Author
-
Tejabhiram Yadavalli, Deepak Shukla, Dinesh Jaishankar, and Neel Thakkar
- Subjects
0301 basic medicine ,Microbiology (medical) ,Chemokine ,Viral pathogenesis ,viruses ,lcsh:Medicine ,Review ,Dengue virus ,medicine.disease_cause ,Endoglycosidase ,Virus ,heparanase ,03 medical and health sciences ,chemistry.chemical_compound ,0302 clinical medicine ,medicine ,Immunology and Allergy ,Heparanase ,Molecular Biology ,General Immunology and Microbiology ,biology ,lcsh:R ,Heparan sulfate ,herpes simplex virus ,Virology ,3. Good health ,Cell biology ,030104 developmental biology ,Infectious Diseases ,Herpes simplex virus ,chemistry ,030220 oncology & carcinogenesis ,biology.protein ,heparan sulfate - Abstract
Heparan sulfate (HS) is ubiquitously expressed on mammalian cells. It is a polysaccharide that binds growth factors, cytokines, and chemokines, and thereby controls several important physiological functions. Ironically, many human pathogens including viruses interact with it for adherence to host cells. HS functions can be regulated by selective modifications and/or selective cleavage of the sugar chains from the cell surface. In mammals, heparanase (HPSE) is the only known enzyme capable of regulating HS functions via a selective endoglycosidase activity that cleaves polymeric HS chains at internal sites. During homeostasis, HPSE expression and its endoglycosidase activity are tightly regulated; however, under stress conditions, including infection, its expression may be upregulated, which could contribute directly to the onset of several disease pathologies. Here we focus on viral infections exemplified by herpes simplex virus, dengue virus, human papillomavirus, respiratory syncytial virus, adenovirus, hepatitis C virus, and porcine respiratory and reproductive syncytial virus to summarize recent advances in understanding the highly significant, but emerging roles, of the enzyme HPSE in viral infection, spread and pathogenesis.
- Published
- 2017
36. Viral Activation of Heparanase Drives Pathogenesis of Herpes Simplex Virus-1
- Author
-
Satvik Hadigal, Alex Agelidis, Deepak Shukla, and Dinesh Jaishankar
- Subjects
0301 basic medicine ,Male ,Viral pathogenesis ,medicine.medical_treatment ,viruses ,Herpesvirus 1, Human ,medicine.disease_cause ,heparanase ,Pathogenesis ,Mice ,0302 clinical medicine ,Interferon ,lcsh:QH301-705.5 ,Glucuronidase ,Mice, Inbred BALB C ,interferon ,Flow Cytometry ,Immunohistochemistry ,3. Good health ,Cytokine ,030220 oncology & carcinogenesis ,Female ,heparan sulfate ,medicine.symptom ,medicine.drug ,Inflammation ,Biology ,General Biochemistry, Genetics and Molecular Biology ,Article ,Cell Line ,03 medical and health sciences ,cornea ,transcription factors ,medicine ,Animals ,Humans ,Heparanase ,Transcription factor ,Wound Healing ,Herpes Simplex ,herpes simplex virus ,cytokines ,ophthalmology ,030104 developmental biology ,Herpes simplex virus ,lcsh:Biology (General) ,Microscopy, Fluorescence ,inflammation ,Immunology ,Virus Activation ,HeLa Cells - Abstract
Summary Herpes simplex virus-1 (HSV-1) causes lifelong recurrent pathologies without a cure. How infection by HSV-1 triggers disease processes, especially in the immune-privileged avascular human cornea, remains a major unresolved puzzle. It has been speculated that a cornea-resident molecule must tip the balance in favor of pro-inflammatory and pro-angiogenic conditions observed with herpetic, as well as non-herpetic, ailments of the cornea. Here, we demonstrate that heparanase (HPSE), a host enzyme, is the molecular trigger for multiple pathologies associated with HSV-1 infection. In human corneal epithelial cells, HSV-1 infection upregulates HPSE in a manner dependent on HSV-1 infected cell protein 34.5. HPSE then relocates to the nucleus to regulate cytokine production, inhibits wound closure, enhances viral spread, and thus generates a toxic local environment. Overall, our findings implicate activated HPSE as a driver of viral pathogenesis and call for further attention to this host protein in infection and other inflammatory disorders.
- Published
- 2017
37. Cultured corneas show dendritic spread and restrict herpes simplex virus infection that is not observed with cultured corneal cells
- Author
-
Tejabhiram Yadavalli, Kyle Mangano, Neel Thakkar, Shrey Patel, Sati Zeynep Tekin, Dinesh Jaishankar, Alex Agelidis, and Deepak Shukla
- Subjects
Gene Expression Regulation, Viral ,0301 basic medicine ,Corneal Infection ,Cell Survival ,viruses ,Biology ,Virus Replication ,Article ,Virus ,Cell Line ,Cornea ,03 medical and health sciences ,Organ Culture Techniques ,Viral entry ,Interferon ,medicine ,Humans ,Simplexvirus ,Cells, Cultured ,Virus Release ,Infectivity ,Multidisciplinary ,Herpes Simplex ,Dendrites ,Virus Internalization ,Virology ,3. Good health ,030104 developmental biology ,medicine.anatomical_structure ,Viral replication ,Cytokines ,sense organs ,Ex vivo ,medicine.drug - Abstract
Herpes simplex virus-1 (HSV-1) causes life-long morbidities in humans. While fever blisters are more common, occasionally the cornea is infected resulting in vision loss. A very intriguing aspect of HSV-1 corneal infection is that the virus spread is normally restricted to only a small fraction of cells on the corneal surface that connect with each other in a dendritic fashion. Here, to develop a comprehensive understanding of the susceptibility of human corneal epithelial (HCE) cells to HSV-1 infection, we infected HCE cells at three different dosages of HSV-1 and measured the outcomes in terms of viral entry, gene and protein expression, viral replication and cytokine induction. In cultured cells, infectivity and cytokine induction were observed even at the minimum viral dosage tested, while a more pronounced dose-restricted infectivity was seen in ex vivo cultures of porcine corneas. Use of fluorescent HSV-1 virions demonstrated a pattern of viral spread ex vivo that mimics clinical findings. We conclude that HCE cell cultures are highly susceptible to infection whereas the cultured corneas demonstrate a higher ability to restrict the infection even in the absence of systemic immune system. The restriction is helped in part by local interferon response and the unique cellular architecture of the cornea.
- Published
- 2017
- Full Text
- View/download PDF
38. 821 Anti-tumor activity induced by topical delivery of HSP70iQ435A
- Author
-
I. C. Le Poole, Dinesh Jaishankar, Emilia R. Dellacecca, Cormac Cosgrove, James P. Mahon, and Steven W. Henning
- Subjects
Antitumor activity ,Cancer research ,Cell Biology ,Dermatology ,Molecular Biology ,Biochemistry - Published
- 2019
- Full Text
- View/download PDF
39. Liposome-Mediated Herpes Simplex Virus Uptake Is Glycoprotein-D Receptor-Independent but Requires Heparan Sulfate
- Author
-
Vaibhav Tiwari, Deepak Shukla, Dinesh Jaishankar, Lorrie Burnham, Kevin S. Jones, and Jeffrey M. Thompson
- Subjects
0301 basic medicine ,Microbiology (medical) ,viruses ,Biology ,medicine.disease_cause ,Microbiology ,03 medical and health sciences ,chemistry.chemical_compound ,0302 clinical medicine ,Viral entry ,medicine ,Cationic liposome ,Original Research ,Transfection ,Heparan sulfate ,virus-cell interactions ,Herpesvirus glycoprotein B ,Molecular biology ,3. Good health ,Oncolytic virus ,030104 developmental biology ,Herpes simplex virus ,chemistry ,Lipofectamine ,030220 oncology & carcinogenesis ,viral entry ,heparan sulfate - Abstract
Cationic liposomes are widely used to facilitate introduction of genetic material into target cells during transfection. This study describes a non-receptor mediated herpes simplex virus type-1 (HSV-1) entry into the Chinese hamster ovary (CHO-K1) cells that naturally lack glycoprotein D (gD)-receptors using a commercially available cationic liposome: lipofectamine. Presence of cell surface heparan sulfate (HS) increased the levels of viral entry indicating a potential role of HS in this mode of entry. Loss of viral entry in the presence of actin de-polymerizing or lysosomotropic agents suggests that this mode of entry results in the endocytosis of the lipofectamine-virus mixture. Enhancement of HSV-1 entry by liposomes was also demonstrated in vivo using a zebrafish embryo model that showed stronger infection in the eyes and other tissues. Our study provides novel insights into gD receptor independent viral entry pathways and can guide new strategies to enhance the delivery of viral gene therapy vectors or oncolytic viruses.
- Published
- 2016
- Full Text
- View/download PDF
40. Extended Release of an Anti-Heparan Sulfate Peptide From a Contact Lens Suppresses Corneal Herpes Simplex Virus-1 Infection
- Author
-
Deepak Shukla, Dinesh Jaishankar, Tibor Valyi-Nagy, Jason S. Buhrman, and Richard A. Gemeinhart
- Subjects
0301 basic medicine ,Ganciclovir ,Male ,Corneal Infection ,Contact Lenses ,Swine ,contact lens ,Eye Infections, Viral ,Herpesvirus 1, Human ,Biology ,medicine.disease_cause ,Ocular herpes ,Cornea ,03 medical and health sciences ,chemistry.chemical_compound ,Mice ,0302 clinical medicine ,medicine ,virus infection ,Animals ,Humans ,Corneal epithelium ,Immunology and Microbiology ,corneal epithelium ,Mice, Inbred BALB C ,Heparan sulfate ,medicine.disease ,Virology ,peptide ,3. Good health ,Contact lens ,Disease Models, Animal ,030104 developmental biology ,medicine.anatomical_structure ,Herpes simplex virus ,chemistry ,Delayed-Action Preparations ,Drug delivery ,030221 ophthalmology & optometry ,Keratitis, Herpetic ,Female ,sense organs ,Heparitin Sulfate ,medicine.drug - Abstract
Herpes simplex virus-1 (HSV-1) is a double-stranded DNA virus widely prevalent in humans and estimated to have infected 60% to 90% of the population.1 Primary and recurrent ocular herpetic infections cause corneal scarring, neovascularization, and epithelium and stromal keratitis; in severe cases, these result in blindness.2,3 While the current available topical treatments include the use of trifluorothymidine (TFT) or the use of ganciclovir gels, oral treatments involve the use of steroids, acyclovir and its analogues valacyclovir that are nucleoside analogues. Dual treatments with steroids are also prescribed by clinicians.4 However, some limitations to the above mentioned treatments include the following: Eye drops constitute ∼90% of ophthalmic medications and have advantages, including ease of administration, safety and efficacy, but this is limited by low residence time due to rapid drainage from the lacrimal ducts resulting in frequent applications5,6; Increasing cases of resistance to acyclovir, the first line of therapy in HSV infections7,8; Use of steroids causes ocular complications such as cataract and steroid-induced glaucoma9–11; and TFT has been shown to pose toxicity at high doses and hence cannot be prescribed for prolonged usage.12,13 While all of these treatments are effective, the limitations pose the requirement of new treatment options for ocular herpes, which may also include improved methods of administration. The envelop glycoprotein D (gD) of HSV-1 is essential for HSV-1 entry and infection.14,15 To date, the following surface receptors have been identified that bind gD and allow infection to proceed: herpesvirus entry mediator (HVEM), Nectin-1 and ‐2, and 3-O-sulfated heparan sulfate (3-OS-HS), a rare modification of the polysaccharide heparan sulfate (HS).15,16 Heparan sulfate is a highly abundant and ubiquitously expressed cell surface receptor, which is also a key molecule for infection as it is required for attachment, viral surfing, capsid penetration, and its removal is needed during egress of newly generated virions.16–18 Therefore, in our quest to target this very common entry coreceptor for antiviral effects, prior studies in our laboratory identified two 12-mer peptides, called G1 and G2, which showed strong anti–HSV-1 properties.19 The peptides G1 and G2 were specifically selected against HS and 3-OS-HS respectively. The in vivo prophylactic activity of the peptides was confirmed using a corneal infection model.19 In another study from our laboratory, the therapeutic effect of G2 along with acyclovir as a potential dual therapy was also tested.20 The aim of this study was to determine the antiviral efficacy of a relatively more stable variant of G2 peptide, G2-C, delivered via contact lenses for extended antiviral therapy. Herpes simplex virus 1 infects corneal epithelium where HS is normally prevalent during initial infection.20 The parental G2 peptide is known to inhibit the interaction19 of virus with HS, but it must be maintained for extended periods to maintain activity. As a potential alternative, many studies have successfully investigated the use of contact lenses for drug delivery to improve the bioavailability and residence time of topically applied drugs.21 Using a similar strategy, we wanted to deliver the G2-C peptide using a vehicle that would release the peptide over an extended period of time, thereby increasing the residence time of the peptide on the corneal epithelium. Our results demonstrate that a commercially available contact lens loaded with the G2-C peptide solution is able to prolong the release of the peptide and the released peptide is able to block HSV-1 infection and replication in natural target cells as well as in corneal infection models.
- Published
- 2016
41. Characterization of a Proteolytically Stable D-Peptide That Suppresses Herpes Simplex Virus 1 Infection: Implications for the Development of Entry-Based Antiviral Therapy
- Author
-
Deepak Shukla, Dinesh Jaishankar, Anita Bogdanov, Abraam M. Yakoub, and Tibor Valyi-Nagy
- Subjects
Proteases ,Proteolysis ,viruses ,Immunology ,Peptide ,Herpesvirus 1, Human ,Biology ,medicine.disease_cause ,Microbiology ,Antiviral Agents ,In vivo ,Virology ,Vaccines and Antiviral Agents ,medicine ,Humans ,Cells, Cultured ,chemistry.chemical_classification ,Oligopeptide ,medicine.diagnostic_test ,Virus Internalization ,medicine.disease ,Entry into host ,Herpes simplex virus ,chemistry ,Insect Science ,Oligopeptides ,Encephalitis ,Peptide Hydrolases - Abstract
Uncontrolled herpes simplex virus 1 (HSV-1) infection can advance to serious conditions, including corneal blindness or fatal encephalitis. Here, we describe a highly potent anti-HSV-1 peptide (DG2) that inhibits HSV-1 entry into host cells and blocks all aspects of infection. Importantly, DG2 is highly resistant to proteases and shows minimal toxicity, paving the way for prophylactic or therapeutic application of the peptide in vivo .
- Published
- 2014
Catalog
Discovery Service for Jio Institute Digital Library
For full access to our library's resources, please sign in.