David Granville

Professor

Research Interests

Proteases
Granzymes
Extracellular matrix
Chronic inflammation
Aging
Disease models
Autoimmune disease
Skin
Vascular biology

Relevant Thesis-Based Degree Programs

Research Options

I am available and interested in collaborations (e.g. clusters, grants).
I am interested in and conduct interdisciplinary research.
 
 

Research Methodology

Cell Biology
Animal models
Human tissue and models
Immunology
Wound healing
Enzymology

Recruitment

Postdoctoral Fellows
Any time / year round

I am looking for post-doctoral fellows. Trainees will be actively involved in the translational research effort pertaining to understanding the physiological and/or pathophysiological role(s) of the serine protease family, granzymes, in the context of skin or vascular inflammation regeneration, in aging, autoimmune and/or chronic disease. Granzymes are a family of 5 serine proteases in humans. With the exception of GzmA and GzmB, very little is known about the other 3 proteases so there are many opportunities to carve out a niche, drug development, and for publications/patents. The job is highly translational and well suited for trainees seeking greater involvement in all steps in taking basic bench research discoveries through to clinical application and potentially commercialization. Our research program spans from basic biochemistry/molecular biology through to target validation, proof-of-concept in animal and human models with a strong connection to industry. Trainees will be responsible for designing and implementing studies to further elucidate mechanisms of disease through the use of in vitro and in vivo models. Dr. Granville’s laboratory (www.granzymes.com) utilizes a variety of disease models related to aging, autoimmunity, injury, inflammation and impaired healing as they pertain to cardiovascular, pulmonary, skin, musculoskeletal, and neuroinflammatory disorders. We also interact heavily with industry and clinicians. 

QUALIFICATIONS:

The candidate should have a solid scientific and technical background with excellent communication, interpersonal and organizational skills. In addition, the candidate will be highly motivated, willing to learn and comfortable with responsibility.

Ph.D. in immunology, molecular & cell biology, experimental pathology, dermatology, experimental medicine, vascular biology or similar discipline.

Expertise and hands-on experience in animal handling and breeding, including all aspects of designing, implementing, analyses, tissue collection, histology pertaining to relevant animal models of disease, is essential.

Background in proteomics, degradomics, transcriptomics would be an asset.

Familiarity with in vivo testing/characterization of small molecule or antibodies in animals, including relevant models of aging, autoimmune and/or chronic disease is required.

Proficiency in cell culture, histology, biochemistry and molecular biology techniques.

Applicants with experience and a strong track record with murine models of aging, autoimmune and/or chronic disease are especially encouraged to apply.

Previous biochemical/cell biology experience with proteases, protease activity assays, extracellular matrix/proteoglycan biology, immunology, histology is a definite asset but we do have experienced people in the lab to teach procedures if necessary.

A proven track record with a minimum of 2-3 first author publications in reputable journals.

Experience with statistical analyses and scientific writing. The candidate must excel in both written and oral communication. Previous experience with grant writing will be considered an asset.

PERSONAL ATTRIBUTES:

Self-starter; Must be highly motivated; Attentive to details and deadlines; Accountable; Flexible; Demanding of excellence; Committed while maintaining balance and perspective; Willing to make and learn from mistakes; Others describe you as honest, trustworthy, positive and respectful, and gets along well with others; a person of high integrity, a good listener, a straightforward communicator, and a team builder; Capable of working independently and in teams; Comfortable with risk and uncertainty; Attention to detail; Excellent vocabulary, oral communication and writing skills; Looking to build something of value; A good sense of humour, social and networking skills are desirable.

I support public scholarship, e.g. through the Public Scholars Initiative, and am available to supervise students and Postdocs interested in collaborating with external partners as part of their research.
I support experiential learning experiences, such as internships and work placements, for my graduate students and Postdocs.
I am open to hosting Visiting International Research Students (non-degree, up to 12 months).
I am interested in hiring Co-op students for research placements.

Complete these steps before you reach out to a faculty member!

Check requirements
  • Familiarize yourself with program requirements. You want to learn as much as possible from the information available to you before you reach out to a faculty member. Be sure to visit the graduate degree program listing and program-specific websites.
  • Check whether the program requires you to seek commitment from a supervisor prior to submitting an application. For some programs this is an essential step while others match successful applicants with faculty members within the first year of study. This is either indicated in the program profile under "Admission Information & Requirements" - "Prepare Application" - "Supervision" or on the program website.
Focus your search
  • Identify specific faculty members who are conducting research in your specific area of interest.
  • Establish that your research interests align with the faculty member’s research interests.
    • Read up on the faculty members in the program and the research being conducted in the department.
    • Familiarize yourself with their work, read their recent publications and past theses/dissertations that they supervised. Be certain that their research is indeed what you are hoping to study.
Make a good impression
  • Compose an error-free and grammatically correct email addressed to your specifically targeted faculty member, and remember to use their correct titles.
    • Do not send non-specific, mass emails to everyone in the department hoping for a match.
    • Address the faculty members by name. Your contact should be genuine rather than generic.
  • Include a brief outline of your academic background, why you are interested in working with the faculty member, and what experience you could bring to the department. The supervision enquiry form guides you with targeted questions. Ensure to craft compelling answers to these questions.
  • Highlight your achievements and why you are a top student. Faculty members receive dozens of requests from prospective students and you may have less than 30 seconds to pique someone’s interest.
  • Demonstrate that you are familiar with their research:
    • Convey the specific ways you are a good fit for the program.
    • Convey the specific ways the program/lab/faculty member is a good fit for the research you are interested in/already conducting.
  • Be enthusiastic, but don’t overdo it.
Attend an information session

G+PS regularly provides virtual sessions that focus on admission requirements and procedures and tips how to improve your application.

 

ADVICE AND INSIGHTS FROM UBC FACULTY ON REACHING OUT TO SUPERVISORS

These videos contain some general advice from faculty across UBC on finding and reaching out to a potential thesis supervisor.

Postdoctoral Fellows

Graduate Student Supervision

Doctoral Student Supervision

Dissertations completed in 2010 or later are listed below. Please note that there is a 6-12 month delay to add the latest dissertations.

Viral Protease Disruption of Host Transcription and Translation Factors in the Pathogenesis of Coxsackievirus B3 Induced Viral Myocarditis (2016)

Myocarditis, inflammation of the heart muscle, is a spectrum of conditions causing significant morbidity and mortality, yet scientific and clinical knowledge related to this entity is limited. One of the most common and best studied causes of myocarditis is infection by coxsackievirus B3 (CVB3). An improved understanding of the science behind CVB3 myocarditis is critical to establishing better diagnostic and therapeutic strategies for affected individuals. CVB3 infection redirects numerous cellular pathways from physiologic processes to viral replication, often mediated by viral proteases. Two viral targets in this process are death associated protein 5 (DAP5) and nuclear pore complex protein 98 (Nup98). DAP5 is a translation initiation factor specific to internal ribosome entry site (IRES) mediated translation. Nup98 is a component of the nuclear pore complex and a transcription factor.In this thesis, I hypothesize that viral proteases contribute to the pathogenesis of viral myocarditis through interaction with DAP5 and Nup98, redirecting translation and transcription towards viral replication.Using in vitro (plasmid expressed viral proteases), in situ (CVB3 infection in cell culture), and in vivo (mouse myocarditis model) models, I demonstrate that viral protease 2A is responsible for the cleavage of DAP5 and Nup98 during CVB3 infection. Both cleavage events I show to be integral to the viral lifecycle using over expression of recombinant fragments and siRNA inhibition of that expression.These results suggest two previously unidentified targets for improved diagnostics and therapeutics for myocarditis, both areas for future research.

View record

The Extracellular role of Granzyme B in Abdominal Aortic Aneurysm (2014)

Background: Abdominal aortic aneurysm (AAA) is an age-related disease characterized by progressivedegradation of elastic lamellae, defective collagen architecture and medial smooth muscle cell loss. We previously demonstrated that knocking out the serine protease granzyme B (GZMB) reduces incidenceand severity of AAA in mice; however, while GZMB is known for its role in apoptosis, it also accumulates extracellularly during inflammation and can cleave extracellular matrix (ECM) components such as decorin and fibrillin-1. We hypothesized that GZMB contributes to AAA development through thedegradation of vascular ECM and that the inhibition of extracellular GZMB would reduce the incidence and severity of AAA progression.Methods: Human aneurysmal samples were obtained and apolipoprotein E(apoE)-knockout (KO),GZMB/apoE-double knockout (GDKO) and perforin/apoE-DKO (PDKO) mice were implanted withosmotic minipumps releasing angiotensin II for 28 days to induce AAA formation. Additional apoE-KOmice were injected with the GZMB inhibitor, serpin A3N (SA3N, 4-120 μg/kg) or anti-GZMB neutralizingantibody (1 mg/kg) prior to pump implantation. Tissues were assessed for aneurysm pathology,inflammation and ECM composition. Collagen content was analysed by second harmonic generation andtransmission electron microscopy.Results: Human aneurysmal tissues showed elevated levels of GZMB immunopositivity compared tocontrols. A significant reduction in AAA incidence and severity was observed in GDKO mice compared toapoE-KO, whereas perforin deficiency was not protective against AAA. A dose-dependent reduction inthe frequency of aortic rupture was observed in mice that received SA3N or anti-GZMB antibodytreatment. Pre-incubation with SA3N prevented decorin cleavage by GZMB in vitro. Reduced GZMB anda corresponding reduction in loss of adventitial decorin were observed in SA3N and anti-GZMB-treatedmice while collagen density was increased. Adventitial collagen from SA3N-treated mice exhibitedsignificantly higher fibre density and reduced fibril size irregularity.Conclusions: GZMB promotes destruction of the elastic lamellae via degradation of fibrillin-1 anddestabilization of elastic microfibrils while GZMB-mediated degradation of decorin contributes to loss ofadventitial collagen organization and density. The extracellular inhibition of GZMB prevented decorinloss and enabled a beneficial remodelling of adventitial collagen in response to medial injury, leading tohigher vessel tensile strength and increased resistance to aortic rupture.

View record

Granzyme B in skin aging, injury and repair (2013)

Granzyme B (GzmB) is a serine protease that can be released into the extracellular spacesby immune cells during chronic inflammation where it is capable of degrading severalcomponents of the extracellular matrix (ECM). Several chronic inflammatory skin diseases havedemonstrated elevated levels of GzmB however the exact role of GzmB in the skin remainspoorly understood. Apolipoprotein E (ApoE) is a protein highly expressed in the skin, where itcan regulate inflammation through its anti-oxidative and anti-inflammatory properties. Micedeficient in ApoE develop an inflammatory skin phenotype when fed a high fat diet indicative ofpremature aging featuring ECM remodeling, hair graying, hair loss and frailty. I thereforehypothesized that GzmB contributes to skin aging, injury and impaired healing in ApoEknockout (ApoE-KO) mice through the degradation of ECM proteins. In the present dissertation,I identified the high fat diet-fed ApoE-KO mouse as a model that displays several characteristicfeatures of skin aging including skin thinning and collagen disorganization. Furtherinvestigations also identified that high fat diet-fed ApoE-KO mice show defects in cutaneouswound healing such as delayed wound closure, reduced contraction and altered collagen content.These changes became worse with age and high fat diet. To test the role of GzmB in this process,GzmB/ApoE double knockout (DKO) mice were generated. These DKO mice were protectedfrom skin thinning and collagen disorganization even when fed a high fat diet, suggesting thatGzmB plays a role in ECM remodeling during aging of the skin in ApoE-KO mice. Furtherinvestigation revealed that GzmB-mediated degradation of the proteoglycan decorin is likely tobe a key mechanism by which GzmB contributes to collagen disorganization and skin aging inApoE-KO mice. Furthermore, DKO mice showed improved wound healing compared to ApoEKOmice featuring faster wound closure, increased contraction and reduced fibronectin degradation. In vitro cleavage assays revealed that fibronectin fragments generated by GzmBmatched those identified in non-healing ApoE-KO mouse wounds. In summary, my findingssuggest that extracellular GzmB contributes to skin aging and impaired healing in ApoE-KOmouse skin through the degradation of ECM components such as decorin and fibronectin.

View record

Granzyme B in vascular remodeling and pathological angiogenesis (2013)

Granzyme B (GZMB) is a serine protease that is expressed by a variety of immune cells and is abundant in a large number of chronic inflammatory disorders. GZMB is highly expressed in cytotoxic lymphocytes where it serves as the main effector molecule of the granule exocytosis pathway by which cytotoxic immune cells mediate target cell death through intracellular delivery of GZMB, leading to activation of apoptotic signaling cascades. GZMB can also accumulate extracellularly during inflammation, where it can cleave a range of extracellular matrix (ECM) proteins that may disrupt cell-matrix interactions and modulate the bioavailability of matrix-bound growth factors. In this dissertation I have explored the intracellular and extracellular roles of GZMB in vascular remodeling in disease. By examining human atherosclerotic plaques, I discovered an imbalance between GZMB and its endogenous inhibitor, proteinase inhibitor 9 (PI-9). PI-9 expression by vascular smooth muscle cells (VSMC) in plaques was reduced with increased disease severity. Elevated levels of GZMB in advanced lesions were correlated with reduced PI-9 expression and increased VSMC apoptosis. These findings suggest that VSMC are more susceptible to GZMB-induced apoptosis in advanced lesions due to reduced PI-9 expression. While examining the extracellular activities of GZMB on vascular remodeling, I focused on the role of GZMB-mediated cleavage of fibronectin (FN), a known GZMB substrate. FN has a major role in regulating angiogenesis as it facilitates endothelial cell (EC) migration and capillary formation, as well as binding to angiogenic growth factors in the ECM including vascular endothelial growth factor (VEGF). VEGF is a potent vascular permeabilizing agent that is sequestered in the ECM by binding FN. GZMB-mediated FN cleavage resulted in reduced EC adhesion, migration and capillary tube formation. In addition, GZMB-mediated FN cleavage induced the release of VEGF from the ECM and promoted VEGF-dependent vascular leakage in vivo. Thus, GZMB may contribute to the progression and/or persistence of chronic inflammation by dysregulating angiogenesis and promoting vascular permeability. Collectively, the results of this work suggest that both intracellular and extracellular GZMB activities contribute to vascular remodeling and pathological angiogenesis.

View record

Extracellular Granzyme B and Pathophysiological Implications (2012)

Granzyme B (GzmB) is a serine protease that contributes to immune-mediated elimination ofcells by initiating a tightly-regulated form of death known as apoptosis. However, duringinflammation, GzmB leaks out and accumulates in the extracellular space, retains its activity,and proficiently cleaves extracellular matrix (ECM) proteins. I therefore hypothesized thatextracellular GzmB is capable of cleaving novel ECM substrates, contributing to dysregulatedECM integrity and function in disease. In the present dissertation I identified eleven novelextracellular GzmB substrates. Further investigations revealed that GzmB-mediatedproteoglycan cleavage was implicated in the dysregulation of active transforming growth factorbeta(TGF-β) sequestration and bioavailability. GzmB cleavage sites were identified in biglycanand betaglycan and active TGF-β was shown to be released from decorin, biglycan andbetaglycan. The pathophysiological role of my findings were further investigated and validatedusing animal models of disesase in which inflammation and elevated GzmB are observed.Evidence of fibrillin-1 and decorin cleavage were observed in atherosclerosis, abdominal aorticaneurysm and in skin aging pathogenesis. I also assessed the activity of GzmB in advancedatherosclerosis using perforin/apolipoprotein E- double knockout (Perf/apoE-DKO) andgranzyme B/apolipoprotein E-double knockout (GzmB/apoE-DKO) mice. Interestingly, unlikeour aneurysm findings whereby only GzmB/apoE-DKO mice were protected, both Perf/apoEDKOand GzmB/apoE-DKO mice were protected from atherosclerosis compared to apoE-KOcontrols, suggesting the intracellular Perf-dependent activities of granzymes are also importantin the pathogenesis of atherosclerosis. In summary, GzmB is a protease that functions bothintracellularly and extracellularly in disease. My findings suggest that the use of Perf knockoutmice alone to study the role of GzmB in disease should be re-evaluated given the increasing evidence in both animal models and in human disease showing elevated GzmB in bodily fluids is associated with inflammation and age.

View record

Master's Student Supervision

Theses completed in 2010 or later are listed below. Please note that there is a 6-12 month delay to add the latest theses.

Granzyme B : a novel therapeutic target for radiation dermatitis (2023)

Radiation dermatitis (RD) is observed in up to 90% of patients receiving radiation therapy. Symptoms of RD include redness, scaling, and crusted wounds. Severe symptoms can prevent future radiation treatments. Current treatments are not effective and may cause severe side effects. Granzyme B (GzmB) was originally characterized as a serine protease secreted by cytotoxic lymphocytes to promote target cell apoptosis. It is now recognized that GzmB can be secreted by other immune cell and non-immune cell populations. GzmB is elevated in a number of inflammatory skin disorders, such as atopic dermatitis, autoimmune blistering, impaired wound healing and scarring. Previous studies suggest that extracellular GzmB cleaves E-cadherin, filaggrin, and decorin leading to reduced epithelial barrier function and scarring. Based on these findings, I hypothesized that GzmB contributes to increased RD severity and delayed healing through the cleavage of epithelial barrier proteins and/or matrix proteins.GzmB expression and cell source, E-cadherin, filaggrin, and decorin levels were assessed using immunohistochemical analysis of biopsies from RD patients. The role of GzmB was investigated in an established RD murine model, comparing GzmB knockout (GzmB-/-) to wild type (WT) mice. The dorsal skin was exposed to a single 40 gray dose of radiation. RD severity was blindly scored. Tissues were harvested at days 4, 14, and 46 post-radiation exposure. Tissues were examined for GzmB, immune cell infiltrate, scarring, and decorin. Macrophages and mast cells expressed GzmB while E-cadherin, filaggrin, and decorin were reduced in human RD skin compared to healthy skin. GzmB-/- mice exhibited a decrease in RD severity compared to WT mice from day 4 to day 12. CD3+ T-cell levels were reduced and neutrophil infiltration was observed at day 14. GzmB was expressed by resident mast cells in WT mice. GzmB-/- mice exhibited less scar area and more decorin than WT mice at day 46. In summary, GzmB may contribute to RD severity through E-cadherin, filaggrin, and decorin cleavage. The RD murine model suggests that depletion of GzmB attenuates severity of RD and could mitigate scarring. These studies provide a rationale for pursuing GzmB as a novel therapeutic target for the treatment of RD.

View record

Granzyme B mediates cleavage of Thrombospondin-2: impact on keratinocyte viability and adhesion (2020)

The epidermis, consisting of keratinocytes, is the first defense against the exterior environmentof the skin. Integral to maintaining the homeostasis of skin function by aidingin thermoregulation, fluid retention, and pathogen defense, the extracellular matrix compositionaids in regulating keratinocyte migration after injury. Impaired healing processes can progressinto chronic non-healing wounds, leading to infection and mortality. Granzyme B (GzmB) is aserine protease, significantly elevated in chronic non-healing wounds. GzmB impairment ofwound healing is attributed to its ability to sustain its proteolytic activity and downstreaminflammation, inhibiting wound repair and tissue remodeling following injury. GzmB isclassically known for its intracellular role in cytotoxic lymphocyte-mediated apoptosis. Recentevidence demonstrates that extracellular GzmB has the ability to cleave a variety of importantextracellular proteins. Thrombospondin-2 (TSP-2) is an extracellular glycoprotein secreted by avariety of cells known to be involved in cell-cell and cell-matrix interactions. TSP-2 has beenreported to impair viability, adhesion, and migration in endothelial cells and squamouscarcinoma cells. Although extensive studies have been done with TSP-2, literature investigatingthe role of TSP-2 in the skin has largely been neglected. I hypothesized that GzmB cleaves TSP2, and GzmB cleavage of TSP-2 mediates extracellular matrix disorganization, alteredkeratinocyte viability, and reduced adhesion. To investigate this hypothesis purposed two aims.In the first aim, cell-free protease assays were utilized to determine whether TSP-2 is aproteolytic substrate of GzmB. In the second aim, the impact of TSP-2-mediated cleavage wasassessed using cultured human keratinocytes. TSP-2 was cleaved by GzmB in a time- and dose-dependent manner. Further, while GzmB-mediated TSP-2 cleavage did not affect cell viability, it did reduce cellular adhesion in a dose-dependent manner. In summary, TSP-2 is a proteolyticsubstrate of GzmB and such cleavage interferes with keratinocyte adhesion.

View record

Granzyme K: a novel contributor in cardiac allograft vasculopathy (2020)

Background - A major factor limiting survival for patients that have undergone cardiac transplantation is cardiac allograft vasculopathy (CAV). CAV is a fibroproliferative inflammatory form of vascular rejection mediated by immune cells and which is initiated upon damage to the graft endothelium and medial smooth muscle cells (SMC). Granzymes are a family of five serine proteases in humans. Granzymes have been shown to exert roles in cell death, endothelial dysfunction, inflammation and matrix remodeling. Granzyme K (GzmK) specifically can promote endothelial dysfunction and the production of inflammatory mediators IL-1β, IL-6, and monocyte chemotactic protein-1. As such, we hypothesized that GzmK contributes to CAV.Methods - An infrarenal aortic interposition graft was completed across a complete major histocompatibility complex (MHC)-mismatch with recipients being either wildtype or GzmK-KO mice. Allografts were then assessed for CAV severity and compared to human CAV samples. The effects of GzmK on human SMC were also assessed in vitro. Given that atherosclerosis shares similar underlying mechanisms to CAV, GzmK deposition was also characterized in atherosclerosis.Results - Human CAV samples demonstrated increased neointimal GzmK deposition as compared to unaffected native coronaries (p= 0.017). GzmK primarily localized to the medial and neointimal layers. Similar deposition patterns were observed in murine transplants. When the role of GzmK was examined, GzmK deficiency resulted in reduced CAV with less neointima formation (p=0.019) and less luminal obstruction (p
View record

Granzyme B disrupts cell-cell adhesion and epithelial barrier function (2018)

The skin is comprised of multiple layers of keratinocytes which together form a barrier to the external environment, regulating temperature, water loss, and pathogen exposure. As such the skin barrier is vital for health as well as disease prevention. Disruption of the epithelial barrier can result in infection, allergen exposure, and inflammation, culminating into severe conditions. Many autoimmune conditions, such as pemphigus, involve a dysregulation and accumulation of immune cells, this results in a disruption in skin barrier causing a loss of function. Granzyme B (GzmB) is a serine protease that is expressed and secreted by a variety of immune and non-immune cells. It can accumulate in the extracellular milieu and retain its proteolytic functions resulting in chronic inflammation and impaired tissue repair due to extracellular matrix (ECM) remodeling. As such, I hypothesized that GzmB disrupts epithelial barrier function through the proteolytic cleavage of cell junction proteins. The present study investigated the impact of GzmB on epithelial barrier dysfunction using Electric Cell-substrate Impedance Sensing (ECIS) and western blot analyses of intercellular junction cleavage fragments. Human formalin fixed, paraffin embedded blistered skin tissue was assessed for the presence of GzmB. GzmB treatment resulted in a loss of E-cadherin staining on the cell membrane which was supported by western blot analysis of the cell supernatants. Additionally, we observed a dose-dependent increase in E-cadherin fragmentation in GzmB-treated cells compared to controls. HaCaT cells exhibited a significant decrease in barrier function when treated with GzmB while cells treated with GzmB in the presence of a specific GzmB inhibitor remained unaffected. While absent in normal skin, GzmB was observed in abundance within the intra-epidermal blister in addition to the surrounding epithelium. In summary, GzmB contributes to a decline in epithelial barrier function in part through the proteolytic cleavage of cell-cell junctions.

View record

Granzyme b: role in spinal cord injury and impeded repair (2018)

The inflammatory cascade following spinal cord injury (SCI) involves multiple cellular and molecular responses that can both aid and impede recovery. A large component of the wound response is the infiltration of immune cells that secrete pro-inflammatory cytokines and proteases. Granzyme B (GzmB) is a serine protease released by immune cells that negatively affects wound healing through its intracellular and extracellular protease activity. GzmB is abundant in neuroinflammatory conditions and contributes to neuron and oligodendrocyte cell death. In this study we investigate the role of GzmB in tissue injury, inflammation and functional recovery following SCI. An SCI was induced in wild-type (WT) and GzmB knockout (GzmB-KO) mice at thoracic level 9. Mouse locomotion was observed over the course of 6 weeks using three behaviour tests (Basso mouse scale, rotarod and horizontal ladder). Lesions were harvested for histological analysis and sections stained with markers for neurons (NeuN) and dyed for myelin (Eriochrome Cyanine). A second cohort of mice were maintained for 1 week after SCI and probed for GzmB expression and cellular localization. GzmB expression was probed using markers for macrophages or microglia (CD68). GzmB-KO mice exhibited significantly improved motor scores, increased myelin and neural survival compared to WT controls. GzmB expression was observed in macrophages at 7 days post injury. In summary, GzmB is elevated and contributes to neurotoxicity, demyelination and impaired functional recovery following SCI.

View record

Extracellular Granzyme K Mediates Endothelial Inflammation Through the Cleavage of Protease Activated Receptor-1 (2016)

Granzymes are a family of serine proteases that were once thought to function exclusively as mediators of cytotoxic lymphocyte-induced target cell death. However, non-lethal roles for granzymes, including Granzyme K (GzK), have been recently proposed. As recent studies have observed elevated levels of GzK in plasma of patients diagnosed with sepsis, we hypothesized that extracellular GzK induces a pro-inflammatory response in endothelial cells. In the present study, extracellular GzK proteolytically activated Protease Activated Receptor-1 (PAR-1) leading to increased IL-6 and MCP-1 production in Human Umbilical Venous Endothelial Cells (HUVEC). Enhanced expression of ICAM-1 along with an increased capacity for adherence of THP-1 cells was also observed. Characterization of downstream pathways implicated the MAPK p38 pathway for ICAM-1 expression, and both the p38 and the ERK1/2 pathways in cytokine production. GzK also increased TNFα–induced inflammatory adhesion molecule expression. Furthermore, the physiological inhibitor of GzK, IαIp, significantly inhibited GzK activity in vitro. In summary, extracellular GzK is not cytotoxic but promotes a pro-inflammatory response in endothelial cells.

View record

Granzyme B Inhibits Keratinocyte Migration by Disrupting Epidermal Growth Factor Receptor (EGFR)-Mediated Signaling (2016)

Chronic skin ulceration is a common complication and cause of morbidity in the elderly, diabetic, obese and/or immobile populations. Effective therapies that adequately promote efficient closure and remodelling of chronic wounds are lacking. Inflammation and excessive protease accumulation and activity are thought to play key roles in the impairment of normal wound healing. Granzyme B (GzmB) is a serine protease that was, until recently, believed to function exclusively in cytotoxic lymphocyte-mediated apoptosis. However, during dysregulated and/or chronic inflammation, GzmB can accumulate in the extracellular milieu, retain its activity, and cleave a number of important extracellular proteins. Epidermal growth factor receptor (EGFR) is a transmembrane receptor involved in cellular processes such as proliferation and migration. EGFR signaling is integral to the wound healing process. I hypothesized that GzmB impairs EGF-induced keratinocyte migration by impairing EGFR signaling. The present study investigated the effects of GzmB on keratinocyte cell migration. Using Electric Cell Substrate Impedance Sensing (ECIS) and other in vitro wound healing assays, the present study demonstrates that GzmB inhibits keratinocyte migration by interfering with the EGFR pathway. GzmB limited cell transition into a migratory morphology and was found to reduce ligand-induced EGFR phosphorylation. Inhibition of GzmB reversed the aforementioned effects in HaCaT cells. In summary, data from the present study suggests a key role for GzmB in the pathogenesis of impaired wound healing through the reduction of EGFR signaling and cell migration.

View record

Serpina3n accelerates wound closure in a murine model of diabetic wound healing (2014)

Chronic, non-healing wounds are a major complication of diabetes and are characterized by chronic inflammation and excessive protease activity. While once thought to function primarily as a pro-apoptotic serine protease, granzyme B (GzmB) can also accumulate in the extracellular matrix during chronic inflammation and cleave extracellular matrix (ECM) proteins that are essential for proper wound healing, including fibronectin. We hypothesized that GzmB contributes to the pathogenesis of impaired diabetic wound healing through excessive degradation of the ECM. In the first part of the thesis, we demonstrated that the majority of GzmB was secreted by mast cells and localized in the wound edges and granulation tissues of completely reepithelialized diabetic mouse wounds at higher levels. Subsequently, we observed that GzmB induced detachment of mouse embryonic fibroblasts and also showed that co-incubation with a mouse serine protease inhibitor, serpina3n (SA3N), abrogated this effect. Finally, we administered SA3N to diabetic mouse wounds and found that wound closure including both reepithelialization and contraction were significantly increased in wounds treated with SA3N. Histological and immunohistochemical analyses of the SA3N-treated wounds revealed a more mature, proliferative granulation tissue phenotype as indicated by increased cells with proliferative activity, vascularization, contractile myofibroblasts, as well as collagen deposition in remodeling tissues. Skin homogenates from SA3N-treated wounds also exhibited greater levels of full-length intact fibronectin when compared to control wounds. In summary, our findings suggested that GzmB contributes to the pathogenesis of diabetic wound healing through the proteolytic cleavage of fibronectin that are essential for normal wound closure, and that inhibition of GzmB can promote granulation tissue maturation and collagen deposition. These results offer preliminary evidence that a GzmB inhibitor may be a relevant therapeutic target in wound management therapy.

View record

Publications

Current Students & Alumni

This is a small sample of students and/or alumni that have been supervised by this researcher. It is not meant as a comprehensive list.
 
 

If this is your researcher profile you can log in to the Faculty & Staff portal to update your details and provide recruitment preferences.

 
 

Sign up for an information session to connect with students, advisors and faculty from across UBC and gain application advice and insight.