• Users Online: 663
  • Home
  • Print this page
  • Email this page
Home About us Editorial board Ahead of print Current issue Search Archives Submit article Instructions Subscribe Contacts Login 


 
 
Table of Contents
REVIEW ARTICLE
Year : 2020  |  Volume : 57  |  Issue : 1  |  Page : 1-13

Severe malaria: Biology, clinical manifestation, pathogenesis and consequences


1 Malaria Research Group, Department of Biosciences and Bioengineering, Indian Institute of Technology, Guwahati, India
2 Institute of Pharmaceutical Research, GLA University, Mathura, Uttar Pradesh, India

Date of Submission30-Jan-2019
Date of Acceptance02-Aug-2019
Date of Web Publication05-Feb-2021

Correspondence Address:
Dr Vishal Trivedi
Malaria Research Group, Department of Biosciences and Bioengineering, Indian Institute of Technology- Guwahati, Guwahati–781 039, Assam
India
Login to access the Email id

Source of Support: None, Conflict of Interest: None


DOI: 10.4103/0972-9062.308793

Rights and Permissions
  Abstract 

Every year, millions of people are infected with malaria, resulting in significant economic losses to the developing and developed nations. The malaria parasite pursues a complicated life cycle in an invertebrate, mosquito and vertebrate host with several distinct stages. In the human host, it invades the liver and red blood cells to complete its life cycle. It is surprising that not only these two organs are under pressure and exhibit functional abnormalities; a large number of clinical studies also support the notion that malaria parasite propagation in the host affects several other organs and modulates functional outcomes of individual cells. Moreover, patients recovered from severe malaria may suffer throughout their life from impairments in organ function such as loss of eyesight, kidney failure, and much more. Thus, malaria infection leads to several pathological outcomes involving different organs and individual cells in the host. The sole purpose of the present article was to give an overview of pathological outcomes during severe malaria along with their molecular mechanisms. A large proportion of deaths associated with disease is contributed by the pathological effect in host due to parasite propagation and toxicity of antimalarials or combination of both. Hence, there is a need, not only to develop antiparasitic agents but also to discover lead molecules to take care of pathophysiological effects in the host. This may help a beginner to get involved with the topic and initiate research work towards improving adjuvant therapy or avoiding serious complications.

Keywords: Anaemia; cerebral malaria; diabetes; host; kidney; malaria


How to cite this article:
Balaji S N, Deshmukh R, Trivedi V. Severe malaria: Biology, clinical manifestation, pathogenesis and consequences. J Vector Borne Dis 2020;57:1-13

How to cite this URL:
Balaji S N, Deshmukh R, Trivedi V. Severe malaria: Biology, clinical manifestation, pathogenesis and consequences. J Vector Borne Dis [serial online] 2020 [cited 2023 Mar 27];57:1-13. Available from: http://www.jvbd.org//text.asp?2020/57/1/1/308793


  Introduction Top


Malaria is a deadly disease which infects over 219 million and kills around 0.4 million people annually[1]. Despite the annual decrease in malaria associated deaths, development of drug resistance in parasites and insecticide resistance in mosquitoes, along with climatic change contribute to the severe outcomes of the disease in developing and developed countries[1],[2],[3]. In humans, malaria is caused by five different species of Plasmodium, namely Plasmodium falciparum, P. malariae, P. ovale, P. vivax and P. knowlesi[4]. Among them, P. falciparum is the most pathogenic species that accounts for 60–70% deaths. Malaria parasite completes its life cycle in two different hosts; invertebrate—Anopheles mosquitoes, and vertebrate—humans [Figure 1].
Figure 1: Life cycle of malaria parasite in vertebrate and invertebrate hosts.

Click here to view


The infection cycle starts with the bite of the Anopheles mosquito (vector) during the sucking of blood from the vertebrate host. In this process, the sporozoites present in the salivary gland of mosquito are being injected, and they quickly migrate towards liver to infect the hepatocytes. Mature merozoites are released from the hepatocytes to blood and each merozoite infects a new red blood cell (RBC) for multiplication[5],[6]. In RBC, merozoites undergo different stages such as rings, trophozoites and schizonts. The mature schizonts stage have 16–24 merozoites which come out of RBC through egress to infect new RBC to start a new cycle. The RBC stages are clinically critical as they are responsible for the symptoms and pathological outcomes in the host during the course of malaria infection[7],[8],[9]. The treatment of severe malaria involves the administration of various anti malarial drugs. But it has been reported that the recovered patients may suffer from deafening, loss of eyesight, kidney failure, etc. depending on the disease severity[1]. The major manifestation of severe malaria comprises of anaemia, acute renal failure, cerebral malaria, pulmonary edema, and/or bleeding.

Multiple organ failures and the cytotoxic effect of antimalarial drugs are the additional challenges in malaria control[10]. It indicates the necessity to develop safe and nontoxic antimalarial medicines, additional adjuvant therapies and also to reduce vector burden on the environment for controlling the disease spread. The present review discusses the impact of malaria on pathophysiology of different organs and its relationship with the appearance of disease conditions in the host. It also focuses on the consequences of severe malaria for better understanding as well as to improve the existing malaria therapies.

Malaria-induced pathology is not limited to one organ

Clinical symptoms of malaria appear during the erythrocytic stage of the malaria parasites. Malarial fever elicits headaches, muscle aches, dry cough, nausea, vomiting, tiredness, etc. along with typical paroxysmal fever [Figure 2]a. A paroxysmal feature of malaria develops with the release of toxic by-products which initiates initial chillness followed by a sudden rise in the body temperature (fever). At advanced stages, manifestations may include severe anaemia, lactic acidosis, hypoxia, splenomegaly, liver diseases, kidney diseases, visual defects, cerebral malaria with neuronal damages, etc. [Figure 2]b. Untreated malaria may lead to coma before death[3],[11],[12]. Giga et al[13] observed anaemia (45%) as the predominant symptom of malaria, followed by convulsions (21%), cerebral malaria (16.4%) and hypotension (11.8%). But Mohanty et al[14] indicate cerebral malaria (52%) as the leading cause of the death in case of P. falciparum infection.
Figure 2: Clinical symptoms and pathology in different organs in host during malaria: (a) Most common symptoms in human during malaria; and (b) Malaria causes damage and affect the physiology of major organs in host.

Click here to view


Earlier, it was believed that the infection caused by P. vivax is clinically mild type and the parasite does not pass in the deep blood vessels of the organs. However, studies from specific geographical area suggest that P. vivax is also associated with severe clinical manifestations and even death[15],[16]. Vivax malaria primarily causes severe anaemia and related problems such as kidney failure, jaundice, respiratory distress, etc[17]. Plasmodium vivax infection (61%) is primarily responsible for the kidney problems in the host[18]. Recently, Mathews et al[19] studied the clinical spectrum of P. vivax infection in 150 patients in the Delhi region of India for 2 yr and observed that about 42% (63 patients) had signs of severe malaria. Plasmodium vivax exclusively resides in reticulocytes and is reported to show higher cytokine production, inflammatory response and endothelial activation as compared to P. falciparum infection. It was suggested that the higher level of CG content in the genome of P. vivax results in higher content of toll-like receptor 9-stimulating CpG motifs causing inflammatory response and cell activation[20].

Another study by Limaye[21] suggested that P. falciparum malaria leads to higher mortality, but differs in malaria pathology when compared with P. vivax. Mixed infection of P. falciparum and P. vivax is more severe than the individual infection. Malaria is also a major cause of death in pregnant women. Postpartum haemorrhage is the major cause of the maternal death during placental malaria[22].

Malaria-mediated pathological outcomes in the host and its mechanism

Plasmodium falciparum infection causes various adverse effects to the host and disturb the body homeostasis. Parasite metabolic wastes damage the host cell and organelle systems such as reticulo-endothelial systems, placenta, visual defects, etc. These effects are schematically depicted in [Figure 2] and discussed in the coming sections.

Severe anaemia

Anaemia is a clinical condition in which haemoglobin (Hb) level falls below 10 g/dl. Severe anaemia is very common during malaria where the level of Hb become <6 g/dl[11], haematocrit <15% in children <12 yr of age. In malaria patients, anaemia develops by following three mechanisms —(a) intravascular haemolysis, (b) increased clearance of malaria infected RBC (iRBC), and (c) decrease in RBC production through dysregulated erythropoiesis [Figure 3]. According to the World Health Organization (WHO), malaria is responsible for about 445,000 deaths per year and anaemia is directly or indirectly responsible for significant proportion of these deaths. Anaemia leads to low level of Hb and contributes to nutritional deficiencies, poor health structure and prevalence of other infectious diseases like bacterial, viral (HIV) and hookworm (intestinal helminth infection)[23],[24]. The incidence of malaria caused especially by P. vivax and P. falciparum is low in American and Asian countries, where the people receive entomological inoculation rate (EIR) of ≤1 per year. Contrastingly, in sub-Saharan Africa region and in low land New Guinea, the EIR has been reported up to 1000 per year. The high incidence of malaria is associated with severe anaemia and results in considerable morbidity and mortality[24].
Figure 3: Malaria affects multiple pathways to cause severe anaemia in host. The toxic by-products from infected RBCs cause rapid lysis or disturbing production of RBCs from bone marrow.

Click here to view


Blood transfusion is one of the primary treatment to combat with severe malarial anaemia. Scott et al[25] estimated that increase in Hb to 1 g/dl can reduce the risk of death by 24% (1.8 million) among children between 28 days to 5 yr. Phytochemicals have also been reported and exhibit antiparasitic activity and lowers severe anaemia in murine malaria. In an in vitro study, ursolice acid, betulinic acid, maslinic acid and oleanolic acid have been shown to have antimalarial activity against chloroquine-sensitive and chloroquine-resistant P. falciparum parasites[26],[27].

Intravascular haemolysis

Intravascular haemolysis refers to the rupture of RBCs within the blood vessels [Figure 3]. During malaria, iRBC lysis releases merozoites which complete their life cycle by infecting fresh RBCs. The cyclic process leads to the reduction in RBCs in the blood and develop anaemia like condition. The iRBC lysis also release Hb derived products such as haemin, methaemoglobin (MetHb), degraded Hb peptides and malarial pigment haemozoin[28]. Metabolites released from iRBC are capable to generate free radicals and cause the oxidative damages to the blood cells. The Hb or its oxidation product MetHb, can damage RBC membrane following a nuclear fission model to enhance the RBC lysis several folds[29]. They also induce the nitric oxide (NO) synthesis by endothelium to contribute into additional oxidative stress. Tumour necrosis factor (TNF)-α is also involved in this process and increases the NO level in blood. Exposure to free radicals leads to the death of RBCs by oxidizing the RBC membrane lipids, proteins and inducing phosphatidylserine (PS) expression on RBC outer membrane[30],[31]. In a mice model, Dey et al[32] reported that the intravascular haemolysis during malaria damages the liver cells. The pro-oxidant molecule (haeme) released from rupture of RBC increases the oxidative load on liver cells, and activated NF-κB increases infiltration of neutrophils, upregulation of chemokines and intercellular adhesion molecules. The severity of haemolysis was positively correlated with the infiltration of the neutrophils and liver damage. Further, the study used deferoxamine (iron chelator) and N-acetylcysteine (anti-oxidant) in reversing the action and preventing the neutrophil infiltration, NF-κB activation and hepatic damage. Non-oxidative stress mediated RBC lysis during malaria is not pursued extensively. Complement mediated haemolysis was reported based on in vitro experiments, but it lacks in vivo support to draw fruitful conclusions[33],[34].

Clearance of red blood cells and infected red blood cells

In malaria, increased RBC clearance rate has been observed [Figure 3]. It may be due to the oxidative stress mediated RBC structural and functional changes, reduced deformability and PS externalization. The life span of RBC is ~120 days in healthy humans[35]. Old RBCs express different age-related markers (ARM) which can be used by macrophages to identify and phagocytose them in spleen, liver and bone marrow[36],[37]. Binding products on RBC surfaces also enhance the macrophagic clearance[36]. It has been reported that during malaria infection RBC expresses markers like rhoptry-associated protein-2 (RSP-2) and phosphatidylserine (PS). Macrophages recognizes the markers on iRBC membrane leading to the macrophagic clearances[36],[38],[39]. Altogether, the markers cause abnormal sequestration of RBCs in spleen and this condition leads to anaemia with reduction in RBC count in the blood.

Dysregulated erythropoiesis

Bone marrow is the prime site of production and maturation of erythropoietic cells. In the process of erythropoiesis, immature RBCs (reticulocytes) in bone marrow further develop and mature into RBC in blood vessels. Spleen and liver are also involved in the erythropoietic process as secondary sites[40]. The interference in erythropoietic process causes a reduction in RBC production, leading to drop in the RBC number in blood to develop anaemia [Figure 3]. Dyserythropoiesis (defective development of RBC) and ineffective erythropoiesis has been observed in human as well as murine Plasmodium infections. A microscopic examination of bone marrow of P. vivax infected patient showed erythroblasts at various stages of degradation[41]. Maggio-Price et al[42] characterized the changes in marrow haematopoietic stem cells in P. berghei-infected rodents. They observed reduction in burst forming unit-erythroid (BFU-E), colony forming unit-erythroid (CFU-E), erythroblasts and bone marrow cellularity in the infected mice after 24 h of infection.

It has been reported that severe malaria causes reduced erythropoiesis and is correlated with increased level of haemozoin, TNF-α, NO, NO-mediated apoptosis in CD34+ cells and reduced level of erythropoietin[43],[44],[45]. Haemozoin, also known as a malaria pigment was once considered as an inert material, but now reported to stimulate immune cells to release inflammatory cytokines. Phagocytosis of haemozoin by the immune cells like macrophages and monocytes results in dysregulation of innate inflammatory mediators. It inhibits the development of erythroid and is also responsible for reduced reticulocyte response[46],[47].

Abnormal level of cytokine causes release of hepcidin in the liver. Hepcidin is a hormone which regulates iron metabolism through its absorption and storage[48],[49]. Hyperhepsidinemia causes the reduction of intestinal iron absorption. On the other hand, hepcidin promotes the iron release from macrophages and leads to accumulation of iron in the liver. This process is known as iron delocalization and it causes hypoferremia to indirectly disturb the erythropoiesis in the bone marrow[50],[51],[52]. Reduction in the level of cytokine RANTES by haemin also impairs the erythropoiesis during malaria[53],[54],[55].

Phagocytosis of iRBC and haemozoin by macrophages causes release of macrophage migration inhibitory factor (MIF), a proinflammatory mediator. During malarial infection, MIF is believed to have a role in bone marrow suppression and inhibition of erythropoietin-dependent erythroid colony formation. Erythropoietic interfering factors during malaria was reviewed in more detail by Pathak and Ghosh[56].

Cerebral malaria

Cerebral malaria (CM) is defined as a serious neurological complication resulting from P. falciparum infection. It is one of the clinical criteria for severe malaria characterised with a Glasgow coma scale (GCS) score of <11 in adults and a Blantyre score of <3 in children. The CM is a leading cause of malaria mortality and it is estimated that CM due to P. falciparum infection is responsible for 20% of adult and 15% of children deaths. The children under the age of 5 yr who reside in high transmission areas have high chances of CM as compared to the adults[57].

Coma is the hallmark symptom during CM along with brain swelling, intracranial hypertension, retinal changes, brainstem signs, bleeding disorders and multi-organ failure[58]. In CM, age is an important factor and it has been observed that children and adults share only few common symptoms at initial time of infection of the disease. The symptoms include fever, convulsions, and neurological abnormalities along with coma[59]. The incidences of CM in children increase the mortality rate despite receiving the malarial chemotherapy. In adults, treatment with intravenous artesunate injection can delay or avoid death[60]. However, patients recovered from CM may suffer with different neurological difficulties such as blindness, ataxia, central hypotonia, cognition, motor function, behavioural changes and epilepsy throughout their life span[61].

The presence of methaemoglobin in the blood stream also potentiate the pathophysiology of CM. The methaemoglobin stimulated endothelium cells exhibit cytoadherence receptors CD36 and intercellular adhesion molecule 1 (ICAM1) on their cell surface which result in adherence of RBCs and contributing to the CM pathophysiology[62]. Post-mortem studies of CM patients showed parasite sequestration in the brain vessels, endothelial injury, blood brain barrier dysfunction and intracranial hypertension[63]. The iRBC sequestration was thought to have a great role in the development of CM. The possible events accountable for CM and associated pathology are given in [Figure 4]. The parasite adhesion protein, P. falciparum erythrocyte membrane protein-1 (PfEMP-1) present on iRBC interacts with ICAM-1 expressed on endothelial cells and allows the adherence of iRBC to the endothelial cell lining[64]. Adhesion of iRBC to endothelial cell promotes further adhesion of iRBC to endothelial lining and agglutination of blood cells in the sequestered area that leads to the blockage of the capillary. The infection of P. falciparum also leads to the activation and accumulation of macrophages, lymphocytes and monocytes at microvasculature. This results in generation of reactive oxygen species (ROS) and secretion of inflammatory cytokines (TNF, IFNγ and lymphotoxin) to produce inflammation and local damage to blood-brain barrier (BBB)[57]. By-products from iRBC also induce RBC aggregation under in vitro conditions and highlight this pathway to explain severe vascular blockage and complication during malaria[31],[65]. Blockage of the capillary develops hypoxia and hypoglycemic condition in the local area which leads to brain cell damage. Lactic acidosis condition further adds up to the cell damage.
Figure 4: The molecular events responsible for pathology of cerebral malaria in host. Malaria parasite sequestration, leaky blood brain barrier, neuronal damages and vesicular blockage are observed during cerebral malaria.

Click here to view


Products released from iRBC causes inflammation and oxidative damages of the endothelium. Haemozoin or iRBC phagocytised macrophages secrete high level of matrix metalloprotease-9 (MMP-9) which can damage the basal lamina around the blood vessels and disrupt the blood brain barrier[46],[66],[67]. The MMPs further enhances the TNF-α and cytokine secretion by macrophages and lymphocytes. In CM, TNF-α, interleukins (IL-12, IL-6 and IL-10) play a critical role in disease development[68]. The role of TNF-α in ICAM-1 overexpression on endothelial surface has been studied in many laboratories. Elevated interleukins and chemokine levels are thought to play critical role in CM[61],[69]. The seizure during CM may be due to the kynurenic metabolites such as quinolinic acid and kynurenic acid. Quinolinic acid is an NMDA receptor agonist and excitotoxin which may have a role in neurological symptoms and complications[70]. Deregulation of Angiopoietin/Tie-2 system was observed in Ugandan children with CM and experimental model of murine[71], but molecular incidences were not explored. There are many unidentified factors involved in the development of CM and the neurological damage is still inconclusive.

The current antimalarial therapy targets specifically to the parasites alone and does not manage the inflammatory cascades. As a result, in spite of treatments, CM is still associated with 20–25% mortality rates. This challenge can be overcome by use of immune-modulators and antioxidants as an adjuvant therapy. Various molecules from synthetic and plant origin have been tested as adjuvant therapy for the management of CM. In instance, curcumin, a major constituent of Curcuma longa has been reported to exhibit antimalarial activity with IC50 value of 5–18 μM. In mice model, where the animal was infected with P. berghei, curcumin-arteether combination therapy prevents the CM and protects the animal from death[70],[72]. Vitamin D was also tested for modulating the proinflammatory response in the mice model infected with P. berghei. The mice supplemented with vitamin D showed a reduced death rate with integrated blood-brain barrier (BBB). The results suggest that vitamin D can be used as a supplement to reduce the severity of CM in malaria endemic regions[73].

Splenomegaly

Abnormal enlargement of the spleen is termed as splenomegaly and is common in severe malaria[11]. During clinical examination, the size of spleen denotes the intensity of the malaria. In malaria-endemic countries, hyper-reactive malarial splenomegaly (HMS) is a major cause of splenomegaly in severe malaria which is characterized by overproduction of IgM antibodies that results in haemolytic anaemia during pregnancy, rupture of spleen and is sometimes fatal. In spite of huge number of literature present on malaria very limited literature deals with HMS[74]. The schematic diagram to explain different biochemical processes leading to splenomegaly during malaria is given in [Figure 5]. Spleen has an important role in filtering the blood and removing aged RBCs, RBC-derived products and bacteria[75]. During malaria, uninfected RBCs and other blood cells undergo abnormal modification (including apoptotic marker expression) which reduces their lifespan. During malaria, immune complexes combine with activated complement C3b which can deposit complement receptor 1 (CR1) present on RBCs. Haemozoin also can enhance the C3b binding on RBC CR1 receptor independently. This event can elicit splenic clearance of uninfected RBC and adds up the work load on spleen leading to splenic infarction[33]. Red pulp macrophages recognize abnormal RBCs, products released from infected RBCs (Hb, MetHb, haemin and haemozoin) and they phagocytose them. The elevated level of cytokines, IgM levels during malaria may also have a role in splenomegaly but the links are not clear[33],[76]. The excessive turnover of RBCs (production and destruction) and toxic by-product released from iRBCs during malaria may lead to spleen dysfunction and splenomegaly.
Figure 5: Malaria infection disturbs spleen function and causes splenomegaly. The parasite propagation within blood induces RBC lysis and accumulation of toxic by-products. The enhanced haemolysis, RBC clearance, red pulp macrophage activation, inflammation and oxidative damages together lead to the splenomegaly in malaria infected host.

Click here to view


Liver complication

About 2.5 to 62% of malaria patients suffer from the liver problems and develop clinical jaundice. The hepatocellular jaundice in severe malaria cases is referred as malarial hepatopathy which is characterized by increased level (about 3 folds than normal upper limit) of serum bilirubin and serum aminotransferases. Malarial hepatopathy is not common in such cases, but it still contributes into malaria mortality up to 2–5%. The schematic diagram to explain different biochemical and molecular pathways leading to liver damage during malaria is given in [Figure 6]. Parasite life cycle starts in the hepatocytes in humans and during this stage they modulate biochemical properties and increase stiffness of the infected hepatocytes[77]. Hepatocytic plasma membrane during sporozoite infection of the liver causes the secretion of the hepatocyte growth factor (HGF) which is known to involve in parasite infection into hepatocytes[78],[79],[80]. Interestingly, there is no remarkable liver pathophysiology during pre-erythrocytic malaria but haemolysis, congestion of hepatocytes, hepatocyte swelling, kupffer cell hyperplasia, deposition of haemozoin, and steatosis have been observed in autopsy of dead malaria patients[81],[82],[83],[84],[85].
Figure 6: The molecular events responsible for liver damage and dysfunction in host during malaria. Malaria parasite mediated RBC lysis and propagation of parasite within liver is responsible for liver damage and dysfunction. Increased detoxification pathways, inflammation and oxidative stress mediated damages together involve in the liver damages during malaria.

Click here to view


During the erythrocytic stage, the free Hb and haemin are being removed by the liver after binding with haptoglobin and haemopexin, respectively[86],[87]. Excess iRBC products oxidatively damage the cells present in microenvironment and lead them to apoptosis or lysis[88],[89]. The RBC released products along with parasitic antigens amplify the immune responses which lead to vascular dysfunction such as blockage, damage, iRBC sequestration, and haemolytic product accumulation in the liver. Accumulation of haemolytic products in the liver, causes severe damage to the hepatocytes through induction of apoptosis involving mitochondrial dysfunction, DNA fragmentation and oxidation of cytoskeleton proteins[32],[90],[91],[92]. Haemozoin also increases the secretion of cytokines from the macrophages and lymphocytes. The elevated levels of NF-κB and TNF-α in kupffer cell generate inflammatory response against hepatocytes during malaria. Hepcidin-mediated iron delocalisation in hepatocytes also damages the hepatocytes[51],[93]. Other mechanisms underlying in liver damage during malaria are still not clear[81],[94].

Guha et al[91] in a mice model reported that the oxidative stress-mediated by free radicals (OH*) was responsible for the death of hepatic cells via apoptosis. Therefore, they suggested that use of antioxidants (NAC, melatonin) and spin trap (TEMPO) can be used as an adjuvant therapy to prevent the mortality in severe malaria. Similarly, in order to improve liver functions and jaundice in severe malaria, ursodeoxycholic acid was also used as an adjunctive therapy along with intravenous artesunate[91],[92],[93],[94],[95].

Renal failure

Severe malaria develops acute kidney injury (AKI) in around 40% malaria patients and responsible for about 75% mortality in the endemic regions. Severe malaria is responsible for functional changes in glomeruli, tubules and interstitial region which arise mainly due to RBC abnormalities. In addition, proteinuria, haematuria, oedema and hypertension have been observed during malaria, especially with P. falciparum infection. During malaria, intravascular haemolysis activates endothelial cells, alter haemodynamics through the generation of oxidative stress, NO and pro-inflammatory cytokine, TNF-α. It leads to renal ischemia, acute tubular necrosis and acute interstitial nephritis[96]. Nephritis during malaria has showed involvement of INF-γ, IL-1α, IL-6 by altering levels of pro-inflammatory and anti-inflammatory cytokines[81],[97],[98]. Elevated level of IL-17 was observed in acute renal failure during malaria[99]. High parasitaemia causes stress to the kidney by various ways. Parasites-mediated lactic acidosis may overload the renal acid-base balance system. The iRBC sequestration and RBC rosetting in the blood vessels of kidney cause hypoxia, which affects the kidney cell activity[100]. IRBC sequestration further allows the deposition of parasites and malaria-related toxic products on the endothelial lining of kidney[101]. It results in endothelial damage as well as immune reactions in the kidneys. Altogether, they direct the kidney failure during malaria[98]. The treatment of kidney disease due to malaria includes use of antimalarial drugs along with fluid replacement and dialysis. In AKI, the association of dialysis in therapy among patients shows better outcome with 25% reduction in mortality and 30% increase in renal recovery[102].

Placental malaria

Placental malaria is another common complication observed in pregnant women infected with P. falciparum [Figure 7]. The compromised immune status in the first and third trimester of pregnancy makes women more susceptible to malaria infections. It had been reported that globally, pregnancy associated malaria (PAM) is accountable for 75,000–200,000 infant deaths per year[103]. Placental malaria leads to miscarriage, poor birth weight, neurological squeal and birth defects in newborn babies in severe cases. Placental malaria risk is higher in women with age below 25 yr and the onset of pregnancy, such as primiparity[104]. In placental malaria, phagocytic cells accumulate at the intervillous space to phagocytose iRBC. The TNF-α and IL-10 activate the macrophage to accumulate haemozoin, iRBC and induces leukocyte infiltration. It results in an increase of the thickness of trophoblast basement membrane to modulate the space between intervillous and perivillous. It results in a reduction of nutrient and oxygen transport across the placenta[105]. The role of elevated C5a during pregnancy may contribute in placental malaria through deregulated angiogenesis, the release of chemokine and cytokine[81]. Placental hormone human chorionic gonadotropin seems to promote malaria parasites.
Figure 7: Malaria infection in pregnant women causes placental complications. Parasitic sequestration in placenta, macrophage mediated inflammatory reactions, accumulation of toxin in intervillus space together effects placental physiology. It affects foetus development and birth outcomes.

Click here to view


Malaria induced diabetes

Malaria mediated damages are not restricted to host organs, but also modulates the host metabolism. The metabolic modulation of the host is observed when there is an increased parasitic metabolism, resulting in altered immune responses and hormonal deregulation. Anaerobic metabolism of parasites directly influences the blood lactic acid level on the host. Parasitaemia level also directly correlates with TNF-α, interleukins and other cytokine level. Hormonal deregulation during malaria is yet unexplained, but the abnormal hormone level has been observed during placental malaria[106]. Elevated blood insulin level (hyperinsulinemia) during severe malaria has been reported in various studies[107],[108].

A recent study about the relation of type-II diabetes with malaria indicated that the incidence of type-II diabetes is increasing among malaria survivors[109]. Interestingly, type-II diabetes also increases the risk of malaria infection. Further, human insulin effects on mosquito indicate that it suppresses the NF-κB level in the mosquito to help the parasite survival in normal or starved mosquitoes[110],[111],[112]. Interestingly, hyperinsulinemia is often connected with hypoglycemia during malaria. During CM, hyperinsulinemia in conjugation with hyperglycemia cause more deaths than hypoglycemic condition alone[113],[114]. These interesting observations need to be explored for the connection between the interaction of insulin with parasites and its down-stream effects in disease development.

Malarial retinopathy

Malarial retinopathy is also one of the important consequences which has been observed in cases of severe malaria. Malarial retinopathy shows the cluster of some unique signs such as whitening of the retina, retinal haemorrhages, changes in ocular blood vessels to orange or white and papilledema. A number of cases of malaria retinopathy was reported in Africa, especially in children where they suffer from CM or severe malarial anaemia (SMA)[115],[116],[117],[118]. Lewallen et al[119] were the first to observe the unusual sign in the retina among the children suffering from CM in Malawi region by direct and indirect ophthalmoscopy. The cytoadherence properties of P. falciparum iRBC were well-established. The histopathology studies of eye show the sequestration of parasite-infected RBC within the ocular blood vessels by cytoadherence. The CM and SMA have been associated with the combination of retinal abnormalities[115]. The mortality rate and coma also correlate with severity of malarial retinopathy in African children suffering from CM, suggesting that the malarial retinopathy is correlated to the pathophysiology of malaria.


  Conclusion Top


Severe malaria is a global health problem in both endemic and non-endemic areas and is responsible for millions of deaths every year. The studies and results described in this review focused on the several pathological outcomes involving different organs and individual cells in the host due to malaria infection. It highlights the need of developing antiparasitic agents as well as discovering molecules to take care of pathophysiological effects in host. A large proportion of deaths associated with the malaria is contributed by the pathological effect in host due to parasite propagation and toxicity of antimalarials or combination of both. The present review will help the researchers to understand the pathophysiology of the malaria infection and its toxic effects on various organs. Moreover, it may also help the researchers to initiate research work towards improving adjuvant therapy or avoiding serious pathology. The work described in this review might lead to new possibilities for therapeutic interventions, which is urgently needed to reduce the morbidity and mortality of malaria.

Ethical statement: Not applicable.

Conflict of interest: None

 
  References Top

1.
World Malaria Report 2018. Geneva: World Health Organization 2019. Available from: https://apps.who.int/iris/bitstream/ handle/10665/275867/9789241565653-eng.pdf?ua = l (Accessed on November 11, 2019).  Back to cited text no. 1
    
2.
Mishra SR, Dhimal M, Guinto RR, Adhikari B, Chu C. Threats to malaria elimination in the Himalayas. Lancet Glob Health 2016; 4(8): e519.  Back to cited text no. 2
    
3.
Plewes K, Turner GDH, Dondorp AM. Pathophysiology, clinical presentation, and treatment of coma and acute kidney injury complicating falciparum malaria. Curr Opin Infect Dis 2018; 31(1): 69–77.  Back to cited text no. 3
    
4.
Mueller I, Zimmerman PA, Reeder JC. Plasmodium malariae and Plasmodium ovale—the “bashful” malaria parasites. Trends Parasitol 2007; 23(6): 278–83.  Back to cited text no. 4
    
5.
Soulard V, Bosson-Vanga H, Lorthiois A, Roucher C, Franetich JF, Zanghi G, et al. Plasmodium falciparum full life cycle and Plasmodium ovale liver stages in humanized mice. Nat Commun 2015; 6: 7690.  Back to cited text no. 5
    
6.
Amino R, Thiberge S, Martin B, Celli S, Shorte S, Frischknecht F, et al. Quantitative imaging of Plasmodium transmission from mosquito to mammal. Nat Med 2006; 12(2): 220–4.  Back to cited text no. 6
    
7.
Angrisano F, Tan YH, Sturm A, McFadden GI, Baum J. Malaria parasite colonisation of the mosquito midgut—placing the Plasmodium ookinete centre stage. Int J Parasitol 2012; 42(6): 519–27.  Back to cited text no. 7
    
8.
Sherman IW. Biochemistry of Plasmodium (malarial parasites). Microbiol Rev 1979; 43(4): 453–95.  Back to cited text no. 8
    
9.
White NJ, Pukrittayakamee S, Hien TT, Faiz MA, Mokuolu OA, Dondorp AM. Malaria. Lancet 2014; 383(9918): 723–35.  Back to cited text no. 9
    
10.
Sarkar PK, Ahluwalia G, Vijayan VK, Talwar A. Critical care aspects of malaria. J Intensive Care Med 2010; 25(2): 93–103.  Back to cited text no. 10
    
11.
Zaki SA, Shanbag P. Atypical manifestations of malaria. Res Rep Trop Med 2011; 2: 9–22.  Back to cited text no. 11
    
12.
Fairhurst RM, Wellems TE. Plasmodium species (malaria). Mandell, Douglas, and Bennett’s Principles and Practice of Infectious Diseases 2010; 2: 3437–62.  Back to cited text no. 12
    
13.
Giha HA, Elghazali G, A-Elgadir TM, A-Elbasit IE, Eltahir EM, Baraka OZ, et al. Clinical pattern of severe Plasmodium falciparum malaria in Sudan in an area characterized by seasonal and unstable malaria transmission. Trans R Soc Trop Med Hyg 2005; 99(4): 243–51.  Back to cited text no. 13
    
14.
Mohanty S, Mishra SK, Pati SS, Pattnaik J, Das BS. Complications and mortality patterns due to Plasmodium falciparum malaria in hospitalized adults and children, Rourkela, Orissa, India. Trans R Soc Trop Med Hyg 2003; 97(1): 69–70.  Back to cited text no. 14
    
15.
Rahimi BA, Thakkinstian A, White NJ, Sirivichayakul C, Dondorp AM, Chokejindachai W. Severe vivax malaria: A systematic review and meta-analysis of clinical studies since 1900. Malar J 2014; 13: 481.  Back to cited text no. 15
    
16.
Naing C, Whittaker MA, Nyunt Wai V, Mak JW. Is Plasmodium vivax malaria a severe malaria?: A systematic review and meta-analysis. PLoS Negl Trop Dis 2014; 8(8): e3071.  Back to cited text no. 16
    
17.
Imtiaz S, Drohlia MF, Nasir K, Hussain M, Ahmad A. Morbidity and mortality associated with Plasmodium vivax and Plasmodium falciparum infection in a tertiary care kidney hospital. Saudi J Kidney Dis Transpl 2015; 26(6): 1169–76.  Back to cited text no. 17
    
18.
Saravu K, Rishikesh K, Parikh CR. Risk factors and outcomes stratified by severity of acute kidney injury in malaria. PLoS One 2014; 9(3): e90419.  Back to cited text no. 18
    
19.
Mathews SE, Bhagwati MM, Agnihotri V. Clinical spectrum of Plasmodium vivax infection, from benign to severe malaria: A tertiary care prospective study in adults from Delhi, India. Trop Parasitol 2019; 9(2): 88–92.  Back to cited text no. 19
    
20.
Dayananda KK, Achur RN, Gowda DC. Epidemiology, drug resistance, and pathophysiology of Plasmodium vivax malaria. J Vector Borne Dis 2018; 55(1): 1–8.  Back to cited text no. 20
    
21.
Limaye CS, Londhey VA, Nabar ST. The study of complications of vivax malaria in comparison with falciparum malaria in Mumbai. J Assoc Physicians India 2012; 60: 15–8.  Back to cited text no. 21
    
22.
Sayinzoga F, Bijlmakers L, van Dillen J, Mivumbi V, Ngabo F, van der Velden K. Maternal death audit in Rwanda 2009–2013: A nationwide facility-based retrospective cohort study. BMJ Open 2016; 6(1): e009734.  Back to cited text no. 22
    
23.
Calis, JC, Phiri K, Faragher B, Brabin BJ, Bates I, Cuevas LE et al. Severe anaemia in Malawian children. N Engl J Med 2008; 358(9): 888–99.  Back to cited text no. 23
    
24.
White NJ. Anaemia and malaria. Malar J 2018; 17(1): 371.  Back to cited text no. 24
    
25.
Scott SP, Chen-Edinboro LP, Caulfield LE, Murray-Kolb LE. The impact of anemia on child mortality: An updated review. Nutrients 2014; 6(12): 5915–32.  Back to cited text no. 25
    
26.
Mavondo GA, Mzingwane ML. Severe malarial anemia (SMA) pathophysiology and the use of phytotherapeutics as treatment options. In Khan J, editor. Current topics in Anemia. IntechOpen 2018. doi: 10.5772/intechopen.70411.  Back to cited text no. 26
    
27.
Rajendran R, Narashimman BS, Trivedi V. Isolation and quantification of antimalarial N-alkylamides from flower-head derived in vitro callus cultures of Spilanthes paniculata. J Biosci Bioeng 2017; 124(1): 99–107.  Back to cited text no. 27
    
28.
Nuchsongsin F, Chotivanich K, Charunwatthana P, Omodeo-Salè F, Taramelli D, Day NP, et al. Effects of malaria heme products on red blood cell deformability. Am J Trop Med Hyg 2007; 77(4): 617–22.  Back to cited text no. 28
    
29.
Balaji SN, Trivedi V. Extracellular methemoglobin mediated early ROS spike triggers osmotic fragility and RBC destruction: An insight into the enhanced hemolysis during malaria. Indian J Clin Biochem 2012; 27(2):178–85.  Back to cited text no. 29
    
30.
Rifkind JM, Mohanty JG, Nagababu E. The pathophysiology of extracellular hemoglobin associated with enhanced oxidative reactions. Front Physiol 2014; 5: 500.  Back to cited text no. 30
    
31.
Balaji SN, Trivedi V. Extracellular methemoglobin primes red blood cell aggregation in malaria: An in vitro mechanistic study. FEBS Lett 2013; 587(4): 350–7.  Back to cited text no. 31
    
32.
Dey S, Bindu S, Goyal M, Pal C, Alam A, Iqbal MS, et al. Impact of intravascular hemolysis in malaria on liver dysfunction: Involvement of hepatic free heme overload, NF-kappaB activation, and neutrophil infiltration. J Biol Chem 2012; 287(32): 26630–46.  Back to cited text no. 32
    
33.
Biryukov S, Stoute JA. Complement activation in malaria: Friend or foe? Trends Mol Med 2014; 20(5): 293–301.  Back to cited text no. 33
    
34.
Ansar W, Bandyopadhyay SM, Chowdhury S, Habib SH, Mandal C. Role of C-reactive protein in complement-mediated hemolysis in Malaria. Glycoconj J 2006; 23(3–4): 233–40.  Back to cited text no. 34
    
35.
Diederich L, Suvorava T, Sansone R, Stevenson Keller TC, Barbarino F, Sutton TR, et al. On the effects of reactive oxygen species and nitric oxide on red blood cell deformability. Front Physiol 2018; 9: 332.  Back to cited text no. 35
    
36.
de Back DZ, Kostova EB, van Kraaij M, van den Berg Timo K, van Bruggen R. Of macrophages and red blood cells; a complex love story. Front Physiol 2014; 5: 9.  Back to cited text no. 36
    
37.
Bratosin D, Mazurier J, Tissier JP, Estaquier J, Huart JJ, Ameisen JC, et al. Cellular and molecular mechanisms of senescent erythrocyte phagocytosis by macrophages: A review. Biochimie 1998; 80(2): 173–95.  Back to cited text no. 37
    
38.
Briglia M. Rossi MA, Faggio C. Eryptosis: Ally or enemy. Curr Med Chem 2017; 24(9): 937–42.  Back to cited text no. 38
    
39.
Awah NW, Troye-Blomberg M, Berzins K, Gysin J. Mechanisms of malarial anaemia: Potential involvement of the Plasmodium falciparum low molecular weight rhoptry-associated proteins. Acta Trop 2009; 112(3): 295–302.  Back to cited text no. 39
    
40.
Moras M, Lefevre SD, Ostuni MA. From erythroblasts to mature red blood cells: Organelle clearance in mammals. Front Physiol 2017; 8: 1076.  Back to cited text no. 40
    
41.
Wickramasinghe S, Looareesuwan S, Nagachinta B, White NJ. Dyserythropoiesis and ineffective erythropoiesis in Plasmodium vivax malaria. Brit J Haematol 1989; 72(1): 91–9.  Back to cited text no. 41
    
42.
Maggio-Price L, Brookoff D, Weiss L. Changes in hematopoietic stem cells in bone marrow of mice with Plasmodium berghei malaria. Blood 1985; 66(5): 1080–5.  Back to cited text no. 42
    
43.
Awandare GA, Kempaiah, P, Ochiel, DO, Piazza P, Keller CC, Perkins DJ. Mechanisms of erythropoiesis inhibition by malarial pigment and malaria-induced proinflammatory mediators in an in vitro model. Am J Hematol 2011; 86(2): 155–62.  Back to cited text no. 43
    
44.
Thawani N, Tam M, Bellemare MJ, Bohle DS, Olivier M, de Souza JB, et al. Plasmodium products contribute to severe malarial anemia by inhibiting erythropoietin-induced proliferation of erythroid precursors. J Infect Dis 2013: 209(1): 140–9.  Back to cited text no. 44
    
45.
Lamikanra AA, Merryweather-Clarke AT, Tipping AJ, Roberts DJ. Distinct mechanisms of inadequate erythropoiesis induced by tumor necrosis factor alpha or malarial pigment. PLoS One 2015; 10(3): e0119836.  Back to cited text no. 45
    
46.
Deshmukh R, Trivedi V. Phagocytic uptake of oxidized heme polymer is highly cytotoxic to macrophages. PLoS One 2014; 9(7): e103706.  Back to cited text no. 46
    
47.
Lamikanra AA, Theron M, Kooij TWA, Roberts DJ. Hemozoin (malarial pigment) directly promotes apoptosis of erythroid precursors. PLoS One 2009; 4(12): e8446.  Back to cited text no. 47
    
48.
Ganz T. Hepcidin, a key regulator of iron metabolism and mediator of anemia of inflammation. Blood 2003; 102(3): 783–8.  Back to cited text no. 48
    
49.
Ganz T, Nemeth E. Hepcidin and iron homeostasis. Biochim Biophys Acta 2012;1823(9): 1434–43.  Back to cited text no. 49
    
50.
Nweneka CV, Doherty CP, Cox S, Prentice A. Iron delocalisation in the pathogenesis of malarial anaemia. Trans R Soc Trop Med Hyg 2010; 104(3): 175–84.  Back to cited text no. 50
    
51.
Spottiswoode N, Duffy PE, Drakesmith H. Iron, anemia and hepcidin in malaria: The importance of iron in pathophysiologic conditions. Front Pharmacol 2014; 5: 125.  Back to cited text no. 51
    
52.
Coffey R, Ganz T. Iron homeostasis: An anthropocentric perspective. J Biol Chem 2017; 292(31): 12727–34.  Back to cited text no. 52
    
53.
Perkins DJ, Were T, Davenport GC, Kempaiah P, Hittner JB, Ong’echa JM. Severe malarial anemia: Innate immunity and pathogenesis. Int J Biol Sci 2011; 7(9): 1427–42.  Back to cited text no. 53
    
54.
Roumenina LT, Rayes J, Lacroix-Desmazes S, Dimitrov JD. Heme: Modulator of plasma systems in hemolytic diseases. Trends Mol Med 2016; 22(3): 200–13.  Back to cited text no. 54
    
55.
Dobbs KR, Embury P, Vulule J, Odada PS, Rosa BA, Mitreva M, et al. Monocyte dysregulation and systemic inflammation during pediatric falciparum malaria. JCI Insight 2017; 2(18): pii: 95352.  Back to cited text no. 55
    
56.
Pathak VA, Ghosh K. Erythropoiesis in malaria infections and factors modifying the erythropoietic response. Anemia 2016; Article ID 9310905.  Back to cited text no. 56
    
57.
Bruneel F. Human cerebral malaria: 2019 mini review. Revue Neurologique 2019; 175(7): 445–50.  Back to cited text no. 57
    
58.
Newton CR, Hien TT, White N. Cerebral malaria. J Neurol Neurosurg Psychiatry 2000; 69(4): 433–41.  Back to cited text no. 58
    
59.
Hawkes M, Elphinstone RE, Conroy AL, Kain KC. Contrasting pediatric and adult cerebral malaria: The role of the endothelial barrier. Virulence 2013; 4(6): 543–55.  Back to cited text no. 59
    
60.
Kovacs SD, Rijken MJ, Stergachis A. Treating severe malaria in pregnancy: A review of the evidence. Drug Saf 2015; 38(2): 165–81.  Back to cited text no. 60
    
61.
Idro R, Marsh K, John CC, Charles, Newton CRJ. Cerebral malaria: Mechanisms of brain injury and strategies for improved neurocognitive outcome. Pediatr Res 2010; 68(4): 267–74.  Back to cited text no. 61
    
62.
Kumar S, Trivedi V. Extracellular methemoglobin promotes cytoadherence of uninfected RBC to endothelial cells: Insight into cerebral malaria pathology. J Cell Biochem 2019; 120(7): 11140–9.  Back to cited text no. 62
    
63.
Taylor TE, Fu WJ, Carr RA, Whitten RO, Mueller JG, Fosiko NG, et al. Differentiating the pathologies of cerebral malaria by postmortem parasite counts. Nat Med 2004; 10(2): 143–5.  Back to cited text no. 63
    
64.
Mehra A, Jerath G, Ramakrishnan V, Trivedi V. Characterization of ICAM-1 biophore to design cytoadherence blocking peptides. J Mol Graph Model 2015; 57: 27–35.  Back to cited text no. 64
    
65.
Li JL, Li K, Guo Y, Zhao Z, Chen LN, Jiang XH, et al. Current advance in cerebral malaria. Zhongguo Zhong Yao Za Zhi 2017; 42(23): 4548–55.  Back to cited text no. 65
    
66.
Polimeni M, Prato M. Host matrix metalloproteinases in cerebral malaria: New kids on the block against blood-brain barrier integrity? Fluids Barriers CNS 2014; 11(1): 1.  Back to cited text no. 66
    
67.
Deshmukh R, Trivedi V. Pro-stimulatory role of methemoglobin in inflammation through hemin oxidation and polymerization. Inflamm Allergy Drug Targets 2013; 12(1): 68–78.  Back to cited text no. 67
    
68.
Milner DA Jr, Whitten RO, Kamiza S, Carr R, Liomba G, Dzamalala C, et al. The systemic pathology of cerebral malaria in African children. Front Cell Infect Microbiol 2014; 4: 104.  Back to cited text no. 68
    
69.
Dunst J, Kamena F, Matuschewski K. Cytokines and chemokines in cerebral malaria pathogenesis. Front Cell Infect Microbiol 2017; 7: 324.  Back to cited text no. 69
    
70.
Jain K, Sood S, Gowthamarajan K. Modulation of cerebral malaria by curcumin as an adjunctive therapy. Braz J Infect Dis 2013; 17(5): 579–91.  Back to cited text no. 70
    
71.
Higgins SJ, Purcell LA, Silver KL, Tran V, Crowley V, Hawkes M, et al. Dysregulation of angiopoietin-1 plays a mechanistic role in the pathogenesis of cerebral malaria. Sci Transl Med 2016; 8(358): 358ra128.  Back to cited text no. 71
    
72.
Vathsala PG, Dende C, Nagaraj VA, Bhattacharya D, Gobardhan Das, Rangarajan PN, et al. Curcumin-arteether combination therapy of Plasmodium berghei-infected mice prevents recrudescence through immunomodulation. PLoS One, 2012; 7(1): e29442.  Back to cited text no. 72
    
73.
Wu B, Du Y, Feng Y, Wang Q, Pang W, Qi Z, et al. Oral administration of vitamin D and importance in prevention of cerebral malaria. Int Immunopharmacol 2018; 64: 356–63.  Back to cited text no. 73
    
74.
Leoni S, Buonfrate D, Angheben A, Gobbi F, Bisoffi Z. The hyper-reactive malarial splenomegaly: A systematic review of the literature. Malar J 2015; 14: 185.  Back to cited text no. 74
    
75.
Mebius RE, Kraal G. Structure and function of the spleen. Nat Rev Immunol 2005; 5(8): 606–16.  Back to cited text no. 75
    
76.
Buffet PA, Safeukui I, Deplaine G, Brousse V, Prendki V, Thellier M, et al. The pathogenesis of Plasmodium falciparum malaria in humans: Insights from splenic physiology. Blood 2011; 117(2): 381–92.  Back to cited text no. 76
    
77.
Eaton P, Zuzarte-Luisb V, Motab MM, Santosc NC, Prudêncio M. Infection by Plasmodium changes shape and stiffness of hepatic cells. Nanomed 2012; 8(1): 17–9.  Back to cited text no. 77
    
78.
Carrolo M, Giordano S, Cabrita-Santos L, Corso S, Vigário AM, Silva S, et al. Hepatocyte growth factor and its receptor are required for malaria infection. Nat Med 2003; 9(11): 1363–9.  Back to cited text no. 78
    
79.
Jin M, Chen Y, He S, Ryan SJ, Hinton DR. Hepatocyte growth factor and its role in the pathogenesis of retinal detachment. Invest Ophthalmol Vis Sci 2004; 45(1): 323–9.  Back to cited text no. 79
    
80.
Kori LD, Valecha N, Anvikar AR. Insights into the early liver stage biology of Plasmodium. J Vector Borne Dis 2018; 55(1): 9–13.  Back to cited text no. 80
    
81.
Autino B, Corbett Y, Castelli F, Taramelli D. Pathogenesis of malaria in tissues and blood. Mediterr J Hematol Infect Dis 2012; 4(1): e2012061.  Back to cited text no. 81
    
82.
Anand AC, Puri P. Jaundice in malaria. J Gastroenterol Hepatol 2005; 20(9): 1322–32.  Back to cited text no. 82
    
83.
Kochar DK, Singh P, Agarwal P, Kochar SK, Pokharna R, Sareen PK. Malarial hepatitis. J Assoc Physicians India 2003; 51: 1069–72.  Back to cited text no. 83
    
84.
Kochar DK, Agarwal P, Kochar SK, Jain R, Rawat N, Pokharna RK, et al. Hepatocyte dysfunction and hepatic encephalopathy in Plasmodium falciparum malaria. QJM 2003; 96(7): 505–12.  Back to cited text no. 84
    
85.
Goncalves LA, Rodo J, Rodrigues-Duarte L, de Moraes LV, Penha-Gonçalves C. HGF secreted by activated kupffer cells induces apoptosis of Plasmodium-infected hepatocytes. Front Immunol 2017; 8: 90.  Back to cited text no. 85
    
86.
Smith A, McCulloh RJ. Hemopexin and haptoglobin: Allies against heme toxicity from hemoglobin not contenders. Front Physiol 2015; 6: 187.  Back to cited text no. 86
    
87.
Belcher JD, Chen C, Nguyen J, Abdulla F, Zhang P, Nguyen H, et al. Haptoglobin and hemopexin inhibit vaso-occlusion and inflammation in murine sickle cell disease: Role of heme oxygenase-1 induction. PLoS One 2018; 13(4): e0196455.  Back to cited text no. 87
    
88.
Pamplona A, Hanscheid T, Epiphanio S, Mota MM, Vigário AM. Cerebral malaria and the hemolysis/methemoglobin/heme hypothesis: Shedding new light on an old disease. Int J Biochem Cell Biol 2009; 41(4): 711–6.  Back to cited text no. 88
    
89.
Totino PR, Magalhães AD, Silva LA, Banic DM, Daniel-Ribeiro CT, Ferreira-da-Cruz Mde F. Apoptosis of non-parasitized red blood cells in malaria: A putative mechanism involved in the pathogenesis of anaemia. Malar J 2010; 9: 350.  Back to cited text no. 89
    
90.
Dey S, Mazumder S, Siddiqui AA, Iqbal MS, Banerjee C, Sarkar S, et al. Association of heme oxygenase 1 with the restoration of liver function after damage in murine malaria by Plasmodium yoelii. Infect Immun 2014; 82(8): 3113–26.  Back to cited text no. 90
    
91.
Guha M, Kumar S, Choubey V, Maity P, Bandyopadhyay U. Apoptosis in liver during malaria: Role of oxidative stress and implication of mitochondrial pathway. Faseb J 2006; 20(8): 1224–6.  Back to cited text no. 91
    
92.
Guha M, Maity P, Choubey V, Mitra K, Reiter RJ, Bandyopadhyay U. Melatonin inhibits free radical-mediated mitochondrial-dependent hepatocyte apoptosis and liver damage induced during malarial infection. J Pineal Res 2007; 43(4): 372–81.  Back to cited text no. 92
    
93.
Deshmukh R, Trivedi V. Methemoglobin exposure produces toxicological effects in macrophages due to multiple ROS spike induced apoptosis. Toxicol In vitro 2013; 27(1): 16–23.  Back to cited text no. 93
    
94.
Viriyavejakul P, Khachonsaksumet V, Punsawad C. Liver changes in severe Plasmodium falciparum malaria: Histopathology, apoptosis and nuclear factor kappa B expression. Malar J 2014; 13(1): 1–10.  Back to cited text no. 94
    
95.
Treeprasertsuk S, Silachamroon U, Krudsood S, Huntrup A, Suwannakudt P, Vannaphan S, et al. Ursodeoxycholic acid and artesunate in the treatment of severe falciparum malaria patients with jaundice. J Gastroenterol Hepatol 2009; 25: 362–8.  Back to cited text no. 95
    
96.
Nguansangiam S, Day NPJ, Hien TT, Mai NTH, Chaisri U, Riganti M, et al. A quantitative ultrastructural study of renal pathology in fatal Plasmodium falciparum malaria. Trop Med Int Health 2007; 12(9): 1037–50.  Back to cited text no. 96
    
97.
Sinniah R, Rui-Mei L, Kara A. Up-regulation of cytokines in glomerulonephritis associated with murine malaria infection. Int J Exp Pathol 1999; 80(2): 87–95.  Back to cited text no. 97
    
98.
Ehrich JH, Eke FU. Malaria-induced renal damage: Facts and myths. Pediatr Nephrol 2007; 22(5): 626–37.  Back to cited text no. 98
    
99.
Herbert F, Tchitchek N, Bansal D, Jacques J, Pathak S, Bécavin C, et al. Evidence of IL-17, IP-10, and IL-10 involvement in multiple-organ dysfunction and IL-17 pathway in acute renal failure associated to Plasmodium falciparum malaria. J Transl Med 2015; 13: 369.  Back to cited text no. 99
    
100.
Terkawi MA, Nishimura M, Furuoka H, Nishikawa Y. Depletion of phagocytic cells during nonlethal Plasmodium yoelii infection causes severe malaria characterized by acute renal failure in mice. Infect Immun 2016; 84(3): 845–55.  Back to cited text no. 100
    
101.
Dumont ME, Dei-Cas E, Maurois P, Slomianny C, Prensier G, Houcke-Lecomte M, et al. Histopathology of the liver and kidney during malaria: Relation to malaria-induced dyslipoproteinemia. Ann Parasitol Hum Comp 1988; 63(3): 171–83.  Back to cited text no. 101
    
102.
Silva GBD Jr, Pinto JR, Barros EJG, Farias GMN, Daher EF. Kidney involvement in malaria: An update. Rev Inst Med Trop Sao Paulo 2017; 59: e53.  Back to cited text no. 102
    
103.
Rogerson SJ, Hviid L, Duffy PE, Leke RF, Taylor DW. Malaria in pregnancy: Pathogenesis and immunity. Lancet Infect Dis 2007; 7(2): 105–17.  Back to cited text no. 103
    
104.
Magistrado P, Salanti A, Tuikue Ndam NG, Mwakalinga SB, Resende M, Dahlbäck M, et al. VAR2CSA expression on the surface of placenta-derived Plasmodium falciparum-infected erythrocytes. J Infect Dis 2008; 198(7): 1071–4.  Back to cited text no. 104
    
105.
Sharma L, Shukla G. Placental malaria: A new insight into the pathophysiology. Front Med (Lausanne) 2017; 4: 117.  Back to cited text no. 105
    
106.
Megnekou R, Tenou S, Bigoga JD, Djontu JC, Medou FM, Lissom A. Placental malaria and modulation of immune and hormonal responses in Cameroonian women. Acta Trop 2015; 147: 23–30.  Back to cited text no. 106
    
107.
Acquah S, Boampong IN, Eghan Jnr BA, Eriksson M. Evidence of insulin resistance in adult uncomplicated malaria: Result of a two-year prospective study. Malar Res Treat 2014; 2014: 136148.  Back to cited text no. 107
    
108.
Eltahir EM, El-Ghazali G, A-Elgadir TME, A-Elbasit IE, Elbashir MI, Giha HA. Raised plasma insulin level and homeostasis model assessment (HOMA) score in cerebral malaria: Evidence for insulin resistance and marker of virulence. Acta Biochimica Polonica 2010; 57(4): 513–20.  Back to cited text no. 108
    
109.
Rohrig G, Maier WA, Seitz HM. Growth-stimulating influence of human chorionic gonadotropin (HCG) on Plasmodium falciparum in vitro. Zentralbl Bakteriol 1999; 289(1): 89–99.  Back to cited text no. 109
    
110.
Pakpour N, Corby-Harris V, Smithers HM, Cheung KW, Riehle MA, Shirley Luckhart S. Ingested human insulin inhibits the mosquito NF-κB-dependentimmune response to Plasmodium falciparum. Infect Immun 2012; 80(6): 2141–9.  Back to cited text no. 110
    
111.
Zhao YO, Kurscheid S, Zhang Y, Liu L, Zhang L, Loeliger K, et al. Enhanced survival of Plasmodium-infected mosquitoes during starvation. PLoS One 2012; 7(7): e40556.  Back to cited text no. 111
    
112.
Danquah, Bedu-Addo G, Mockenhaupt FP. Type 2 diabetes mellitus and increased risk for malaria infection. Emerg Infect Dis 2010; 16(10): 1601–4.  Back to cited text no. 112
    
113.
Giha H, Elghazali G, A-Elgadir TM, A-Elbasit IE, Elbashir MI. Severe malaria in an unstable setting: Clinical and laboratory correlates of cerebral malaria and severe malarial anemia and a paradigm for a simplified severity scoring. Eur J Clin Microbiol Infect Dis 2009; 28(6): 661–5.  Back to cited text no. 113
    
114.
Tombe M, Bhatt KM, Obel AO. Clinical surprises and challenges of severe malaria at Kenyatta National Hospital, Kenya. East Afr Med J 1993; 70(2): 117–9.  Back to cited text no. 114
    
115.
Maude RJ, Dondorp AM, Sayeed AA, Day Nicholas PJ, White NJ, Beare Nicholas AV. The eye in cerebral malaria: What can it teach us? Trans R Soc Trop Med Hyg 2009; 103(7): 661–4.  Back to cited text no. 115
    
116.
Nanfack CN, Bilong Y, Kagmeni G, Nathan NN, Bella LA. Malarial retinopathy in adult: A case report. Pan Afr Med J 2017; 27: 224.  Back to cited text no. 116
    
117.
Wiwanitkit V. Malarial retinopathy: The summary on contemporaneous hypothesis. Med Hypothesis Discov Innov Ophthalmol 2012; 1(3): 63–4.  Back to cited text no. 117
    
118.
Beare NA, Taylor T, Harding SP, Lewallen S, Molyneux ME. Malarial retinopathy: A newly established diagnostic sign in severe malaria. Am J Trop Med Hyg 2006; 75(5): 790–7.  Back to cited text no. 118
    
119.
Lewallen S, Taylor T. The eyes have it-or do they? Am J Trop Med Hyg. 2017; 96(5): 1007–8.  Back to cited text no. 119
    


    Figures

  [Figure 1], [Figure 2], [Figure 3], [Figure 4], [Figure 5], [Figure 6], [Figure 7]


This article has been cited by
1 Antiplasmodial Activity of Phyllanthus amarus and Associated Impact on Surrogate Markers of Insulin Sensitivity and Resistance Indices
Adefunke O. Opajobi, Ugochukwu E. Uzuegbu, Chinwendu O. Elu, Pere-Ebi Y. Toloyai, Ikechukwu Igwenyi, Innocent Onyesom
Journal of Herbs, Spices & Medicinal Plants. 2022; : 1
[Pubmed] | [DOI]
2 Key factors predicting suspected severe malaria case management and health outcomes: an operational study in the Democratic Republic of the Congo
Jean Okitawutshu, Aita Signorell, Jean-Claude Kalenga, Eric Mukomena, Giulia Delvento, Christian Burri, Fatou Mwaluke, Valentina Buj, Moulaye Sangare, Sylvie Luketa, Nina Brunner, Tristan Lee, Manuel Hetzel, Christian Lengeler, Antoinette Tshefu
Malaria Journal. 2022; 21(1)
[Pubmed] | [DOI]
3 Neutrophils in malaria: A double-edged sword role
Kehinde Adebayo Babatunde, Oluwadamilola Fatimat Adenuga
Frontiers in Immunology. 2022; 13
[Pubmed] | [DOI]
4 A Retrospective Review on Severe Malaria in Colombia, 2007–2020
Jaime Carmona-Fonseca, Mario J. Olivera, María F. Yasnot-Acosta
Pathogens. 2022; 11(8): 893
[Pubmed] | [DOI]
5 Major Achievements in the Design of Quadruplex-Interactive Small Molecules
Eduarda Mendes, Israa M. Aljnadi, Bárbara Bahls, Bruno L. Victor, Alexandra Paulo
Pharmaceuticals. 2022; 15(3): 300
[Pubmed] | [DOI]
6 Blood glucose, insulin and glycogen profiles in Sprague-Dawley rats co-infected with Plasmodium berghei ANKA and Trichinella zimbabwensis
Pretty Murambiwa, Achasih Quinta Nkemzi, Samson Mukaratirwa
PeerJ. 2022; 10: e13713
[Pubmed] | [DOI]



 

Top
 
  Search
 
    Similar in PUBMED
   Search Pubmed for
   Search in Google Scholar for
 Related articles
    Access Statistics
    Email Alert *
    Add to My List *
* Registration required (free)  

 
  In this article
Abstract
Introduction
Conclusion
References
Article Figures

 Article Access Statistics
    Viewed9641    
    Printed91    
    Emailed0    
    PDF Downloaded1381    
    Comments [Add]    
    Cited by others 6    

Recommend this journal